1
|
Ren Y, Liu F, Chen X, Zhang X, Zhao B, Wan Y, Lan Y, Li X, Yang W, Zhu X, Guo Y. Decitabine-containing conditioning improved outcomes for children with higher-risk myelodysplastic syndrome undergoing allogeneic hematopoietic stem cell transplantation. Ann Hematol 2024; 103:1345-1351. [PMID: 38316642 DOI: 10.1007/s00277-024-05628-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2023] [Accepted: 01/13/2024] [Indexed: 02/07/2024]
Abstract
Myelodysplastic syndrome (MDS) is a rare clonal hematopoietic disorder in children. The risk stratification system and treatment strategy for adults are unfit for children. The role of hypomethylating agents (HMAs) in higher-risk childhood MDS has not been identified. This study aimed to investigate the outcomes of hematopoietic stem cell transplantation (HSCT) in children with higher-risk MDS at one single center. A retrospective study was conducted in children with higher-risk MDS undergoing HSCT between September 2019 and March 2023 at Blood Diseases Hospital CAMS. The clinical characteristics and transplantation information were reviewed and analyzed. A total of 27 patients were analyzed, including 11 with MDS with excess blasts (MDS-EB), 14 with MDS-EB in transformation (MDS-EBt) or acute myeloid leukemia with myelodysplasia-related changes (AML-MRC), and 2 with therapy-related MDS/AML (t-MDS/AML). Eight patients harbored monosomy 7. Before transplantation, induction therapy was administered to 25 patients, and 19 of them achieved bone marrow blasts <5% before HSCT. The stem cell source was unmanipulated-related bone marrow or peripheral blood stem cells for nineteen patients and unrelated cord blood for eight. All patients received decitabine-containing and Bu/Cy-based myeloablative conditioning; 26 patients achieved initial engraftment. The cumulative incidences of grade II-IV and grade III-IV acute graft-versus-host disease (GvHD) at 100 days were 65.4% and 42.3%, respectively. The incidence of cGvHD was 38.5%. The median follow-up was 26 (range 4-49) months after transplantation. By the end of follow-up, two patients died of complications and two died of disease progression. The probability of 3-year overall survival (OS) was 84.8% (95%CI, 71.1 to 98.5%). In summary, decitabine-containing myeloablative conditioning resulted in excellent outcomes for children with higher-risk MDS undergoing allogeneic HSCT.
Collapse
Affiliation(s)
- Yuanyuan Ren
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China
- Tianjin Institutes of Health Science, Tianjin, 301600, China
| | - Fang Liu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China
- Tianjin Institutes of Health Science, Tianjin, 301600, China
| | - Xia Chen
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China
- Tianjin Institutes of Health Science, Tianjin, 301600, China
| | - Xiaoyan Zhang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China
- Tianjin Institutes of Health Science, Tianjin, 301600, China
| | - Beibei Zhao
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China
- Tianjin Institutes of Health Science, Tianjin, 301600, China
| | - Yang Wan
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China
- Tianjin Institutes of Health Science, Tianjin, 301600, China
| | - Yang Lan
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China
- Tianjin Institutes of Health Science, Tianjin, 301600, China
| | - Xiaolan Li
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China
- Tianjin Institutes of Health Science, Tianjin, 301600, China
| | - Wenyu Yang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China
- Tianjin Institutes of Health Science, Tianjin, 301600, China
| | - Xiaofan Zhu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China
- Tianjin Institutes of Health Science, Tianjin, 301600, China
| | - Ye Guo
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China.
- Tianjin Institutes of Health Science, Tianjin, 301600, China.
| |
Collapse
|
2
|
Epimutations and Their Effect on Chromatin Organization: Exciting Avenues for Cancer Treatment. Cancers (Basel) 2022; 15:cancers15010215. [PMID: 36612210 PMCID: PMC9818548 DOI: 10.3390/cancers15010215] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 12/14/2022] [Accepted: 12/28/2022] [Indexed: 12/31/2022] Open
Abstract
The three-dimensional architecture of genomes is complex. It is organized as fibers, loops, and domains that form high-order structures. By using different chromosome conformation techniques, the complex relationship between transcription and genome organization in the three-dimensional organization of genomes has been deciphered. Epigenetic changes, such as DNA methylation and histone modification, are the hallmark of cancers. Tumor initiation, progression, and metastasis are linked to these epigenetic modifications. Epigenetic inhibitors can reverse these altered modifications. A number of epigenetic inhibitors have been approved by FDA that target DNA methylation and histone modification. This review discusses the techniques involved in studying the three-dimensional organization of genomes, DNA methylation and histone modification, epigenetic deregulation in cancer, and epigenetic therapies targeting the tumor.
Collapse
|
3
|
Zhang D, Chen J. Efficacy of decitabine combined with allogeneic hematopoietic stem cell transplantation in the treatment of recurrent and refractory acute myeloid leukemia (AML): A systematic review and meta-analysis. Medicine (Baltimore) 2022; 101:e30644. [PMID: 36123842 PMCID: PMC9478241 DOI: 10.1097/md.0000000000030644] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2022] [Accepted: 08/18/2022] [Indexed: 11/29/2022] Open
Abstract
BACKGROUND This analysis aimed to assess the effect of decitabine combined with allogeneic hematopoietic stem cell transplantation (allo-HSCT) in treating recurrent and refractory acute myeloid leukemia. METHOD The present analysis was carried out according to the principles of Preferred Reporting Items for Systematic Reviews and Meta-Analyses guideline statement. Web of Science, Embase, PubMed, The Cochrane Library, CNKI, VIP, and WanFang Data databases were searched for trials published from their corresponding inception to September 13, 2021. Retrospective research or published randomized controlled trials in Chinese or English were ruled out. The methodological quality of the included studies was assessed using the Physiotherapy Evidence Database scale. Mean differences with 95% confidence intervals were used to analyze continuous data. The I2 test was used to determine heterogeneity, and the meta-analysis was conducted using Revman 5.4. RESULTS Eight studies including 795 participants in total were identified. Decitabine and allo-HSCT showed significant reductions in recurrence after transplantation (odds ratio [OR] = 0.29, 95% confidence interval [CI] (0.17, 0.50), P < .00001), leukemia-free survival (OR = 2.17, 95% CI (1.47, 3.21), P < .0001), graft related death (OR = 0.50, 95% CI (0.25, 0.98), P = .04), and significant improvements in complete remission (OR = 0.39, 95% CI = 0.23-0.68, P = .0007) and partial remission (OR = 0.46, 95%CI = 0.27-0.78, P = .004). The median follow-up time, acute graft-versus-host disease, and no remission had no significant difference between treatment and control groups (the median follow-up time: OR = -1.76, 95% CI (-6.28, 2.76), P = .45; acute graft-versus-host disease: OR = 0.72, 95% CI (0.50, 1.03), P = .08; no remission: OR = 3.19, 95%CI = 2.06-4.94, P = .05). Overall, the magnitude of the effect was found to be in the small to moderate range. CONCLUSION Decitabine combined with allo-HSCT can obtain lower recurrence risk and longer disease-free survival time, and improve the prognosis of patients. The safety is relatively stable. Due to the varying quality level of the included studies, the validation of multiple high-quality studies still needs improvement.
Collapse
Affiliation(s)
- Donghui Zhang
- Department of Pharmacy, Gansu Provincial Hospital, Lanzhou, Gansu, China
| | - Jiahui Chen
- Department of Clinical Skills Training Center, Clinical Academic Affairs Office, The First Hospital of Lanzhou University, Lanzhou, China
| |
Collapse
|
4
|
Comparative analysis of Decitabine intensified BUCY2 and BUCY2 conditioning regimen for high-risk MDS patients undergoing allogeneic hematopoietic stem cell transplantation. Bone Marrow Transplant 2022; 57:1063-1071. [PMID: 35459877 DOI: 10.1038/s41409-022-01645-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2021] [Revised: 03/01/2022] [Accepted: 03/15/2022] [Indexed: 11/08/2022]
Abstract
The optimal conditioning regimen for high-risk myelodysplastic syndrome (MDS) patients undergoing allogeneic hematopoietic stem cell transplantation (allo-HSCT) remains elusive. This study aimed to explore the anti-leukemic efficacy and toxicity of Decitabine (Dec, 20 mg/m2/day, day -11 to -7) intensified BUCY2 vs. traditional regimen in high-risk MDS population. We retrospectively evaluated 93 consecutive high-risk MDS patients undergoing allo-HSCT in our institution, comparing discrepancies in clinical characteristics and outcomes between cases using Dec-intensified BUCY2 (n = 52) and traditional BUCY2 regimen (n = 41). Three-year cumulative incidence of relapse after Dec-intensified BUCY2 conditioning was remarkably lower than that of patients using BUCY2 regimen (20.2% vs. 39.0%, p = 0.034). Overall survival and disease-free survival at 3 years for Dec-intensified BUCY2 group were 70.2% and 64.9%, respectively, which were significantly improved when compared with BUCY2 group (51.1% and 43.9%, p = 0.031 and p = 0.027). Furthermore, overall survival and disease-free survival for MDS cases receiving cytoreduction therapy were dramatically better than patients in non-cytoreduction group (p = 0.041, p = 0.047). In summary, the Dec-intensified conditioning regimen could be effective and feasible, providing prominent recurrence control with moderate toxicity for high-risk MDS patients. These patients might also benefit from pre-transplant cytoreductive therapeutic schedules. Larger randomized controlled trials are still needed to further confirm these conclusions.
Collapse
|
5
|
Ko H, Ahn HJ, Kim YI. Methylation and mutation of the inhibin‑α gene in human melanoma cells and regulation of PTEN expression and AKT/PI3K signaling by a demethylating agent. Oncol Rep 2021; 47:37. [PMID: 34958114 DOI: 10.3892/or.2021.8248] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Accepted: 11/23/2021] [Indexed: 11/06/2022] Open
Abstract
Inhibin suppresses the pituitary secretion of follicle‑stimulating hormone and has been reported to act as a tumor suppressor gene in the gonad in mice. Epigenetic modifications, mutations, changes in the loss of heterozygosity (LOH) of the inhibin‑α gene and regulation of gene expression in response to a demethylating agent [5‑aza‑2'‑deoxycytidine (5‑Aza‑dC)] in human melanoma cells were assessed. In addition, the association between a mutation in the 5'‑untranslated region (5'‑UTR) of the inhibin‑α subunit and the expression of phosphatidylinositol 3,4,5‑trisphosphate‑dependent Rac exchanger 2 (PREX2) and phosphatase and tensin homolog (PTEN) as well as AKT/PI3K signaling was determined. The methylation status of the CpG sites of the inhibin‑α promoter was analyzed by methylation‑specific PCR in bisulfite‑treated DNA. Cell viability was counted using the trypan blue assay, mRNA expression was examined via reverse transcription‑quantitative PCR, and protein expression was examined via western blot analysis. The inhibin‑α promoter was hypermethylated in G361, SK‑MEL‑3, SK‑MEL‑24 and SK‑MEL‑28 cells and moderately methylated in SK‑MEL‑5 cells. Inhibin‑α gene mutations were observed in the 5'‑UTR exon 1 of G361, SK‑MEL‑5, SK‑MEL‑24 and SK‑MEL‑28 cells as well as in exon 2 of SK‑MEL‑3 cells. Allelic imbalance, including LOH, in the inhibin‑α gene was detected in human melanoma cells. Treatment with 5‑Aza‑dC increased inhibin‑α mRNA and protein levels, inhibited cell proliferation, and delayed the doubling times of surviving melanoma cells. In 5‑Aza‑dC‑treated cells, PREX2 protein expression was slightly increased in G361 and SK‑MEL‑24 cells and decreased in SK‑MEL3, SK‑MEL‑5 and SK‑MEL‑28 cells. However, the protein expression of PTEN was decreased in melanoma cells. In addition, AKT and PI3K protein phosphorylation levels increased in all melanoma cells, except of G361 cells, demonstrating decreased PI3K protein phosphorylation. These data provided evidence that methylation, mutation and LOH are observed in the inhibin α‑subunit gene and gene locus in human melanoma cells. Furthermore, the demethylating agent reactivated inhibin‑α gene expression and regulated PREX2 expression. AKT/PI3K signaling increased as PTEN expression decreased. In addition, mutations in the tumor suppressor inhibin‑α, PTEN and p53 genes were not associated with transcriptional silencing, gene expression and cell growth as analyzed through experiments and literature reviews. These data demonstrated that methylation and mutations were associated with the inhibin‑α gene in human melanoma cells and indicated the regulation of PTEN expression and AKT/PI3K signaling by a demethylating agent.
Collapse
Affiliation(s)
- Hyunmin Ko
- Department of Surgery, College of Medicine, Kyung Hee University, Dongdaemun, Seoul 02447, Republic of Korea
| | - Hyung Joon Ahn
- Department of Surgery, College of Medicine, Kyung Hee University, Dongdaemun, Seoul 02447, Republic of Korea
| | - Young Il Kim
- Medical Science Research Institute, Kyung Hee University Medical Center, Dongdaemun, Seoul 02447, Republic of Korea
| |
Collapse
|
6
|
Xiao W, Zhou Q, Wen X, Wang R, Liu R, Wang T, Shi J, Hu Y, Hou J. Small-Molecule Inhibitors Overcome Epigenetic Reprogramming for Cancer Therapy. Front Pharmacol 2021; 12:702360. [PMID: 34603017 PMCID: PMC8484527 DOI: 10.3389/fphar.2021.702360] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Accepted: 08/02/2021] [Indexed: 12/12/2022] Open
Abstract
Cancer treatment is a significant challenge for the global health system, although various pharmacological and therapeutic discoveries have been made. It has been widely established that cancer is associated with epigenetic modification, which is reversible and becomes an attractive target for drug development. Adding chemical groups to the DNA backbone and modifying histone proteins impart distinct characteristics on chromatin architecture. This process is mediated by various enzymes modifying chromatin structures to achieve the diversity of epigenetic space and the intricacy in gene expression files. After decades of effort, epigenetic modification has represented the hallmarks of different cancer types, and the enzymes involved in this process have provided novel targets for antitumor therapy development. Epigenetic drugs show significant effects on both preclinical and clinical studies in which the target development and research offer a promising direction for cancer therapy. Here, we summarize the different types of epigenetic enzymes which target corresponding protein domains, emphasize DNA methylation, histone modifications, and microRNA-mediated cooperation with epigenetic modification, and highlight recent achievements in developing targets for epigenetic inhibitor therapy. This article reviews current anticancer small-molecule inhibitors targeting epigenetic modified enzymes and displays their performances in different stages of clinical trials. Future studies are further needed to address their off-target effects and cytotoxicity to improve their clinical translation.
Collapse
Affiliation(s)
- Wenjing Xiao
- School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, China.,Department of Pharmacy, The General Hospital of Western Theater Command of PLA, Chengdu, China
| | - Qiaodan Zhou
- Department of Ultrasonic, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Xudong Wen
- Department of Gastroenterology and Hepatology, Chengdu First People's Hospital, Chengdu, China
| | - Rui Wang
- Information Department of Medical Security Center, The General Hospital of Western Theater Command of PLA, Chengdu, China
| | - Ruijie Liu
- School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, China
| | - Tingting Wang
- School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, China
| | - Jianyou Shi
- Personalized Drug Therapy Key Laboratory of Sichuan Province, Department of Pharmacy, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Yonghe Hu
- School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, China.,Department of Pharmacy, The General Hospital of Western Theater Command of PLA, Chengdu, China
| | - Jun Hou
- School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, China.,Department of Pharmacy, The General Hospital of Western Theater Command of PLA, Chengdu, China
| |
Collapse
|
7
|
Gao C, Qiao T, Yuan S, Zhuang X. The Preliminary Study for Postoperative Radiotherapy Survival Associated with RUNX3 and TLR9 Expression in Lung Cancer. Cancer Manag Res 2021; 13:4497-4507. [PMID: 34113175 PMCID: PMC8186941 DOI: 10.2147/cmar.s305452] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Accepted: 04/23/2021] [Indexed: 11/23/2022] Open
Abstract
Background Many studies have reported that the inflammatory immune response related to TLR9 signaling activation participates in tumor development and affects the treatment outcome. RUNX3 functions as a tumor suppressor by regulating DNA methylation. RUNX3 protein plays an important role in TGF-β signaling pathway that is involved in tumor growth inhibition and apoptosis. At present, radiotherapy is still an important treatment in lung cancer, which induces immune response and affects the therapeutic outcome. The role of TLR9 signaling activation and RUNX3 in this process is not clear. Methods In this study, we investigated the expression of TLR9 in tumor and RUNX3 in surrounding tissues by immunohistochemical methods and analyzed the relationship on postoperative survival in lung cancer. Results We found that the high expression of TLR9 was the risk factor in postoperative survival of lung cancer with no difference in lifetime. The high expression of RUNX3 in lung cancer with TLR9 signaling activation was in favor of progression-free survival and overall survival in postoperative radiotherapy. It suggested that RUNX3 played an important role in lung cancer radiotherapy. In order to determine the effect of RUNX3 in lung cancer radiation with TLR9 signaling activation, we introduced 5-Aza-2ʹ-deoxycytidine (5-Aza-CdR) and exposed lung cancer A459 cells repeatedly. The high expression of RUNX3 especially RUNX3-B in cells treated with 5-Aza-CdR was observed. We examined that 5-Aza-CdR induced more cell blocking in G2/M phase in combining irradiation. Conclusion The result implied that it was feasible to improve radiosensitivity of lung cancer with TLR9 signaling activation by increasing RUNX3 expression, and 5-Aza-CdR was an option in this process.
Collapse
Affiliation(s)
- Caixia Gao
- Jinshan Hospital Center for Tumor Diagnosis & Therapy, Shanghai, 201508, People's Republic of China
| | - Tiankui Qiao
- Jinshan Hospital Center for Tumor Diagnosis & Therapy, Shanghai, 201508, People's Republic of China
| | - Sujuan Yuan
- Jinshan Hospital Center for Tumor Diagnosis & Therapy, Shanghai, 201508, People's Republic of China
| | - Xibing Zhuang
- Jinshan Hospital Center for Tumor Diagnosis & Therapy, Shanghai, 201508, People's Republic of China
| |
Collapse
|
8
|
Low-dose decitabine as part of a modified Bu-Cy conditioning regimen improves survival in AML patients with active disease undergoing allogeneic hematopoietic stem cell transplantation. Bone Marrow Transplant 2021; 56:1674-1682. [PMID: 33637882 DOI: 10.1038/s41409-021-01238-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 01/14/2021] [Accepted: 02/02/2021] [Indexed: 11/08/2022]
Abstract
Relapse is the major cause of mortality in patients with acute myeloid leukemia (AML) after allogeneic hematopoietic stem cell transplantation (allo-HSCT). Effective preventive intervention in high-risk AML may be crucial. In this study, we investigated the clinical efficacy and safety of low dose decitabine (DAC) as part of a modified Busulfan-Cyclophosphamide (Bu-Cy) regimen for high-risk AML patients undergoing allo-HSCT to reduce relapse rate. Fifty-nine patients received DAC (20 mg/m2/d, i.v.) for 5 days, followed by modified Bu-Cy (DAC group). A matched-pair control (CON) group of 177 patients (matched 1:3) received modified Bu-Cy only. The differences were more substantial among patients with active disease: 2-year OS, 80.7% (DAC) versus 43.5% (CON), P = 0.011 and 2-year LFS, 64.9% (DAC) versus 39.2% (CON), P = 0.024. Median time to relapse was 8 months (DAC) versus 5 months (CON) for the entire groups and 6.5 months (DAC) versus 3.5 months (CON) for patients with active disease. In summary, our data indicated that the conditioning regimen containing low dose DAC may confer a survival advantage in high-risk AML patients with active disease undergoing allo-HSCT, and a prospective randomized trial is warranted to confirm these observations.
Collapse
|
9
|
Mpakou V, Spathis A, Bouhla A, Mpazani E, Papageorgiou S, Gkontopoulos K, Glezou E, Thomopoulos T, Foukas P, Pappa V. Synergistic inhibitory effects of low-dose decitabine in combination with bortezomib in the AML cell line Kasumi-1. Exp Ther Med 2021; 21:195. [PMID: 33488804 PMCID: PMC7812574 DOI: 10.3892/etm.2021.9628] [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: 01/20/2020] [Accepted: 11/17/2020] [Indexed: 11/29/2022] Open
Abstract
In the present study, the ability of the proteasome inhibitor bortezomib (BZ), an oxidative stress-inducing agent, to sensitize acute myeloid leukemia (AML) cells to decitabine (Dacogen®, DAC; a DNA methyltransferase inhibitor), in terms of cell viability and differentiation, was investigated. Kasumi-1 AML (M2) cells were treated with low-dose DAC (10, 50, 100, 200 or 400 nΜ), with or without BZ (10 nM). Apoptosis and the cell cycle were evaluated after 24 h of treatment through fluorescence-assisted cell sorting (FACS) with Annexin V/propidium iodide and DAPI staining, respectively. The expression levels of CD193, CD11b, CD13, CD14, CD15, CD16 and CD117 surface differentiation markers were evaluated by FACS after 6 days of treatment. The results indicated significant alterations in cell death and cell cycle phases in Kasumi-1 cells following DAC and BZ combination treatment compared to untreated cells and cells with single treatments. Low-dose DAC/BZ combinations significantly enhanced apoptosis and decreased the population of live Kasumi-1 cells, with 100 and 200 nM of DAC and 10 nM BZ appearing to have the most potent synergistic effect according to a combination index. Furthermore, cell cycle profiling revealed that DAC/BZ treatment synergistically led to G0/G1- and G2/M-phase arrest. By contrast, DAC appeared to promote monocytic and granulocytic differentiation of Kasumi-1 cells more effectively alone than in combination with BZ. BZ acted synergistically with low-dose DAC in vitro, leading to enhanced apoptosis and G0/G1- and G2/M-phase arrest in AML cells, hence prohibiting either DNA synthesis or mitosis. Although further in vivo investigation is necessary, these results provide a strong rationale for the implementation of a combination treatment with DAC and bortezomib in AML therapy, followed by DAC alone, which may achieve better clinical responses and possibly partially overcome the frequently encountered DAC resistance of patients with AML.
Collapse
Affiliation(s)
- Vassiliki Mpakou
- Second Department of Internal Medicine and Research Institute, National and Kapodistrian University of Athens, Attikon University General Hospital, Athens 12462, Greece
| | - Aris Spathis
- Second Department of Pathology, National and Kapodistrian University of Athens, Attikon University General Hospital, Athens 12462, Greece
| | - Anthi Bouhla
- Second Department of Internal Medicine and Research Institute, National and Kapodistrian University of Athens, Attikon University General Hospital, Athens 12462, Greece
| | - Efthimia Mpazani
- Second Department of Internal Medicine and Research Institute, National and Kapodistrian University of Athens, Attikon University General Hospital, Athens 12462, Greece
| | - Sotirios Papageorgiou
- Second Department of Internal Medicine and Research Institute, National and Kapodistrian University of Athens, Attikon University General Hospital, Athens 12462, Greece
| | - Konstantinos Gkontopoulos
- Second Department of Internal Medicine and Research Institute, National and Kapodistrian University of Athens, Attikon University General Hospital, Athens 12462, Greece
| | - Eirini Glezou
- Second Department of Internal Medicine and Research Institute, National and Kapodistrian University of Athens, Attikon University General Hospital, Athens 12462, Greece
| | - Thomas Thomopoulos
- Second Department of Internal Medicine and Research Institute, National and Kapodistrian University of Athens, Attikon University General Hospital, Athens 12462, Greece
| | - Periklis Foukas
- Second Department of Pathology, National and Kapodistrian University of Athens, Attikon University General Hospital, Athens 12462, Greece
| | - Vasiliki Pappa
- Second Department of Internal Medicine and Research Institute, National and Kapodistrian University of Athens, Attikon University General Hospital, Athens 12462, Greece
| |
Collapse
|
10
|
Méndez-Blanco C, Fondevila F, Fernández-Palanca P, García-Palomo A, van Pelt J, Verslype C, González-Gallego J, Mauriz JL. Stabilization of Hypoxia-Inducible Factors and BNIP3 Promoter Methylation Contribute to Acquired Sorafenib Resistance in Human Hepatocarcinoma Cells. Cancers (Basel) 2019; 11:E1984. [PMID: 31835431 PMCID: PMC6966438 DOI: 10.3390/cancers11121984] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 11/27/2019] [Accepted: 12/06/2019] [Indexed: 01/19/2023] Open
Abstract
Despite sorafenib effectiveness against advanced hepatocarcinoma (HCC), long-term exposure to antiangiogenic drugs leads to hypoxic microenvironment, a key contributor to chemoresistance acquisition. We aimed to study the role of hypoxia in the development of sorafenib resistance in a human HCC in vitro model employing the HCC line HepG2 and two variants with acquired sorafenib resistance, HepG2S1 and HepG2S3, and CoCl2 as hypoximimetic. Resistant cells exhibited a faster proliferative rate and hypoxia adaptive mechanisms, linked to the increased protein levels and nuclear translocation of hypoxia-inducible factors (HIFs). HIF-1α and HIF-2α overexpression was detected even under normoxia through a deregulation of its degradation mechanisms. Proapoptotic markers expression and subG1 population decreased significantly in HepG2S1 and HepG2S3, suggesting evasion of sorafenib-mediated cell death. HIF-1α and HIF-2α knockdown diminished resistant cells viability, relating HIFs overexpression with its prosurvival ability. Additionally, epigenetic silencing of Bcl-2 interacting protein 3 (BNIP3) was observed in sorafenib resistant cells under hypoxia. Demethylation of BNIP3 promoter, but not histone acetylation, restored BNIP3 expression, driving resistant cells' death. Altogether, our results highlight the involvement of HIFs overexpression and BNIP3 methylation-dependent knockdown in the development of sorafenib resistance in HCC. Targeting both prosurvival mechanisms could overcome chemoresistance and improve future therapeutic approaches.
Collapse
Affiliation(s)
- Carolina Méndez-Blanco
- Institute of Biomedicine (IBIOMED), University of León, Campus of Vegazana s/n, 24071 León, Spain; (C.M.-B.); (F.F.); (P.F.-P.); (J.G.-G.)
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, Av. de Monforte de Lemos, 5, 28029 Madrid, Spain
| | - Flavia Fondevila
- Institute of Biomedicine (IBIOMED), University of León, Campus of Vegazana s/n, 24071 León, Spain; (C.M.-B.); (F.F.); (P.F.-P.); (J.G.-G.)
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, Av. de Monforte de Lemos, 5, 28029 Madrid, Spain
| | - Paula Fernández-Palanca
- Institute of Biomedicine (IBIOMED), University of León, Campus of Vegazana s/n, 24071 León, Spain; (C.M.-B.); (F.F.); (P.F.-P.); (J.G.-G.)
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, Av. de Monforte de Lemos, 5, 28029 Madrid, Spain
| | - Andrés García-Palomo
- Service of Oncology, Complejo Asistencial Universitario de León, Calle Altos de Nava, s/n, 24001 León, Spain;
| | - Jos van Pelt
- Laboratory of Clinical Digestive Oncology, Department of Oncology, KU Leuven and University Hospitals Leuven and Leuven Cancer Institute (LKI), 3000 Leuven, Belgium; (J.v.P.)
| | - Chris Verslype
- Laboratory of Clinical Digestive Oncology, Department of Oncology, KU Leuven and University Hospitals Leuven and Leuven Cancer Institute (LKI), 3000 Leuven, Belgium; (J.v.P.)
| | - Javier González-Gallego
- Institute of Biomedicine (IBIOMED), University of León, Campus of Vegazana s/n, 24071 León, Spain; (C.M.-B.); (F.F.); (P.F.-P.); (J.G.-G.)
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, Av. de Monforte de Lemos, 5, 28029 Madrid, Spain
| | - José L. Mauriz
- Institute of Biomedicine (IBIOMED), University of León, Campus of Vegazana s/n, 24071 León, Spain; (C.M.-B.); (F.F.); (P.F.-P.); (J.G.-G.)
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, Av. de Monforte de Lemos, 5, 28029 Madrid, Spain
| |
Collapse
|
11
|
Gouveia HJCB, Manhães-de-Castro R, Costa-de-Santana BJR, Mendonça CR, Albuquerque G, Visco DB, Lacerda DC, Toscano AE. Maternal exposure to busulfan reduces the cell number in the somatosensory cortex associated with delayed somatic and reflex maturation in neonatal rats. J Chem Neuroanat 2019; 103:101710. [PMID: 31706849 DOI: 10.1016/j.jchemneu.2019.101710] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Revised: 11/01/2019] [Accepted: 11/01/2019] [Indexed: 01/31/2023]
Abstract
Busulfan is a bifunctional alkylating agent used for myeloablative conditioning and in the treatment of chronic myeloid leukemia due to its ability to cause DNA damage. However, in rodent experiments, busulfan presented a potential teratogenic and cytotoxic effect. Studies have evaluated the effects of busulfan on fetuses after administration in pregnancy or directly on pups during the lactation period. There are no studies on the effects of busulfan administration during pregnancy on offspring development after birth. We investigated the effects of busulfan on somatic and reflex development and encephalic morphology in young rats after exposure in pregnancy. The pregnant rats were exposed to busulfan (10 mg/kg, intraperitoneal) during the early developmental stage (days 12-14 of the gestational period). After birth, we evaluated the somatic growth, maturation of physical features and reflex-ontogeny during the lactation period. We also assessed the effects of busulfan on encephalic weight and cortical morphometry at 28 days of postnatal life. As a result, busulfan-induced pathological changes included: microcephaly, evaluated by the reduction of cranial axes, delay in reflex maturation and physical features, as well as a decrease in the morphometric parameters of somatosensory and motor cortex. Thus, these results suggest that the administration of a DNA alkylating agent, such as busulfan, during the gestational period can cause damage to the central nervous system in the pups throughout their postnatal development.
Collapse
Affiliation(s)
- Henrique J C B Gouveia
- Post Graduate Program in Nutrition, Health Sciences Center, Federal University of Pernambuco, Recife, Pernambuco, Brazil
| | | | - Bárbara J R Costa-de-Santana
- Post Graduate Program in Neuropsychiatry and Behavioral Sciences, Health Sciences Center, Federal University of Pernambuco, Recife, Pernambuco, Brazil
| | - Carolina R Mendonça
- Post Graduate Program in Nutrition, Health Sciences Center, Federal University of Pernambuco, Recife, Pernambuco, Brazil
| | - Glayciele Albuquerque
- Post Graduate Program in Physiotherapy, Health Sciences Center, Federal University of Pernambuco, Recife, Pernambuco, Brazil
| | - Diego Bulcão Visco
- Post Graduate Program in Nutrition, Health Sciences Center, Federal University of Pernambuco, Recife, Pernambuco, Brazil
| | - Diego Cabral Lacerda
- Post Graduate Program in Nutrition, Health Sciences Center, Federal University of Pernambuco, Recife, Pernambuco, Brazil
| | - Ana Elisa Toscano
- Department of Nursing, CAV, Federal University of Pernambuco, Vitória de Santo Antão, Pernambuco, Brazil.
| |
Collapse
|
12
|
Valdez BC, Tang X, Li Y, Murray D, Liu Y, Popat U, Champlin RE, Andersson BS. Epigenetic modification enhances the cytotoxicity of busulfan and4-hydroperoxycyclophosphamide in AML cells. Exp Hematol 2018; 67:49-59.e1. [PMID: 30102945 DOI: 10.1016/j.exphem.2018.08.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Revised: 07/23/2018] [Accepted: 08/05/2018] [Indexed: 01/09/2023]
Abstract
The combination of the DNA-alkylating agents busulfan (Bu) and cyclophosphamide is the most commonly used myeloablative pretransplantation conditioning therapy for myeloid leukemias. However, it is associated with significant nonrelapse mortality, which prohibits dose escalation to control relapse. We hypothesized that combining these two drugs with an epigenetic modifier would increase antileukemic efficacy without jeopardizing patient safety. A preclinical study was performed to determine the synergistic cytotoxicity of Bu, 4-hydroperoxycyclophosphamide (4HC), and the hypomethylating agent decitabine (DAC) in human acute myeloid leukemia (AML) cell lines. Exposure of KBM3/Bu2506 (P53-null) and OCI-AML3 (P53-wild-type) cells to Bu+4HC inhibited cell proliferation by ∼35-39%; addition of DAC increased the inhibition to ∼60-62%. The observed synergistic interactions correlated with DNA damage response activation, increased the production of reactive oxygen species, and decreased mitochondrial membrane potential, release of mitochondrial proapoptotic proteins into the cytoplasm, and induction of caspase-dependent programmed cell death. The Bu+4HC+DAC combination further caused chromatin trapping of DNMT1 with a concomitant increase in DNA damage. In contrast, FMS-like tyrosine kinase 3 internal tandem duplications (FLT3-ITD)-positive AML cell lines were not sensitized to Bu+4HC by inclusion of DAC; addition of the FLT3 kinase inhibitor sorafenib sensitized the FLT3-ITD-positive MV4-11 and MOLM13 cell lines to the triple drug combination by inhibiting the FLT3 signal transduction pathway. Our results therefore provide a rationale for the development of personalized conditioning therapy for patients with P53-mutated and FLT3-ITD-positive AML.
Collapse
Affiliation(s)
- Benigno C Valdez
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas M.D. Anderson Cancer Center, Houston, TX, USA.
| | - Xiaowen Tang
- Department of Hematology, The First Affiliated Hospital of Soochow University, Jiangsu Institute of Hematology, Suzhou, China
| | - Yang Li
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| | - David Murray
- Department of Experimental Oncology, Cross Cancer Institute, Edmonton, Alberta, Canada T6G 1Z2
| | - Yan Liu
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| | - Uday Popat
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| | - Richard E Champlin
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| | - Borje S Andersson
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
13
|
Differential effects of histone deacetylase inhibitors on cellular drug transporters and their implications for using epigenetic modifiers in combination chemotherapy. Oncotarget 2018; 7:63829-63838. [PMID: 27564097 PMCID: PMC5325407 DOI: 10.18632/oncotarget.11561] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2016] [Accepted: 08/11/2016] [Indexed: 02/02/2023] Open
Abstract
HDAC inhibitors, DNA alkylators and nucleoside analogs are effective components of combination chemotherapy. To determine a possible mechanism of their synergism, we analyzed the effects of HDAC inhibitors on the expression of drug transporters which export DNA alkylators. Exposure of PEER lymphoma T-cells to 15 nM romidepsin (Rom) resulted in 40%-50% reduction in mRNA for the drug transporter MRP1 and up to ~500-fold increase in the MDR1 mRNA within 32-48 hrs. MRP1 protein levels concomitantly decreased while MDR1 increased. Other HDAC inhibitors - panobinostat, belinostat and suberoylanilide hydroxamic acid (SAHA) - had similar effects on these transporters. The protein level of MRP1 correlated with cellular resistance to busulfan and chlorambucil, and Rom exposure sensitized cells to these DNA alkylators. The decrease in MRP1 correlated with decreased cellular drug export activity, and increased level of MDR1 correlated with increased export of daunorubicin. A similar decrease in the level of MRP1 protein, and increase in MDR1, were observed when mononuclear cells derived from patients with T-cell malignancies were exposed to Rom. Decreased MRP1 and increased MDR1 expressions were also observed in blood mononuclear cells from lymphoma patients who received SAHA-containing chemotherapy in a clinical trial. This inhibitory effect of HDAC inhibitors on the expression of MRP1 suggests that their synergism with DNA alkylating agents is partly due to decreased efflux of these alkylators. Our results further imply the possibility of antagonistic effects when HDAC inhibitors are combined with anthracyclines and other MDR1 drug ligands in chemotherapy.
Collapse
|
14
|
Li N, Chen Q, Gu J, Li S, Zhao G, Wang W, Wang Z, Wang X. Synergistic inhibitory effects of deferasirox in combination with decitabine on leukemia cell lines SKM-1, THP-1, and K-562. Oncotarget 2018; 8:36517-36530. [PMID: 28388554 PMCID: PMC5482673 DOI: 10.18632/oncotarget.16583] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2016] [Accepted: 03/14/2017] [Indexed: 12/17/2022] Open
Abstract
A multi-center study from the French Myelodysplastic Syndrome (MDS) Group confirmed that iron chelation therapy is an independent prognostic factor that can increase the survival rate of patients who are suffering from transfusion-dependent low-risk MDS. In this study, we aimed to explore this clinical phenomena in vitro, by exploring the synergistic effect of the iron chelator Deferasirox (DFX) and the DNA methyl transferase inhibitor Decitabine (DAC) in the leukemia cell lines SKM-1, THP-1, and K-562. Treatment with both DFX or DAC promoted apoptosis, induced cell cycle arrest, and inhibited proliferation in all three of these cell lines. The combination of DFX and DAC was much greater than the effect of using either drug alone. DFX showed a synergistic effect with DAC on cell apoptosis in all three cell lines and on cell cycle arrest at the G0/G1 phase in K-562 cells. DFX decreased the ROS levels to varying degrees. In contrast, DAC increased ROS levels and an increase in ROS was also noted when the two drugs were used in combination. Treatment of cells with DAC induced re-expression of ABAT, APAF-1, FADD, HJV, and SMPD3, presumably through demethylation. However the combination of DAC and DFX just had strong synergistic effect on the re-expression of HJV.
Collapse
Affiliation(s)
- Nianyi Li
- Department of Haematology, Huashan Hospital, Fudan University, Shanghai, China
| | - Qinfen Chen
- Department of Haematology, Huashan Hospital, Fudan University, Shanghai, China
| | - Jingwen Gu
- Department of Haematology, Huashan Hospital, Fudan University, Shanghai, China
| | - Shuang Li
- Department of Haematology, Huashan Hospital, Fudan University, Shanghai, China
| | - Guangjie Zhao
- Department of Haematology, Huashan Hospital, Fudan University, Shanghai, China
| | - Wei Wang
- Department of Haematology, Huashan Hospital, Fudan University, Shanghai, China
| | - Zhicheng Wang
- Department of Haematology, Huashan Hospital, Fudan University, Shanghai, China
| | - Xiaoqin Wang
- Department of Haematology, Huashan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
15
|
Effects of Decitabine on the proliferation of K562 cells and the expression of DR4 gene. Saudi J Biol Sci 2017; 25:242-247. [PMID: 29472772 PMCID: PMC5816003 DOI: 10.1016/j.sjbs.2017.11.036] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Revised: 11/09/2017] [Accepted: 11/12/2017] [Indexed: 12/26/2022] Open
Abstract
Objective To investigate the role of DR4 gene in the occurrence, development and prognosis of acute myeloid leukemia (AML), find a new regulatory gene of Decitabine for the treatment of AML, namely DR4 gene, and explore the molecular mechanism of AML in the treatment of AML. Methods The methylation level and the mRNA expression level of DR4 gene promoters of bone marrow mononuclear cells in 122 patients with newly diagnosed AML and 24 patients with iron deficiency anemia (IDA) were detected using Methylation specific PCR (MS-PCR) and Q-RT-PCR, respectively, and a correlation analysis of them was conducted. The effects of Decitabine on the proliferation of K562 cells were detected using CCK-8 assay. Then, the effects of Decitabine on the methylation level and the mRNA expression level of DR4 genes of K562 cells treated with Decitabine were detected using MS-PCR and Q-RT-PCR, respectively. The effects of Decitabine on the cell cycle and apoptosis of K562 cells were detected using flow cytometry. Results Compared with the control group, the methylation level (P = .002) of DR4 genes of bone marrow mononuclear cells in patients with newly diagnosed AML was high. The methylation level (P = .01) of DR4 genes of bone marrow mononuclear cells in patients of the positive group of enlargement of liver, spleen and lymph node was lower than that of the negative group, and the methylation level (P = .006) of DR4 genes in patients of the high risk group of clinical stage was lower than that of the low risk group, and the methylation level (P = .03) of DR4 genes in patients of the group where patients did not achieve complete remission (CR1) after a course of induction chemotherapy was lower than that of the group where patients achieved complete remission (CR1) after a course of induction chemotherapy. There was a significant negative correlation (P < .01) between the methylation level and the mRNA expression level of DR4 genes of bone marrow mononuclear cells in 122 patients with newly diagnosed AML. After the K562 cells were treated with Decitabine for 48 h, the methylation level of DR4 gene promoters gradually decreased, while the mRNA expression level of DR4 genes gradually increased, both of which showed a concentration-dependent relationship. After the K562 cells were treated with 5 µmol/L Decitabine for 48 h, the K562 cells in G0/G1 phase and G2/M phase increased significantly, and the K562 cells in S phase decreased significantly. Conclusion DR4 gene played an important role in the occurrence and development of AML. Decitabine can effectively inhibit the proliferation of K562 cells, which probably partly because it can terminate the methylation effect of DR4 gene promoters and restore the mRNA expression of DR4 genes.
Collapse
|
16
|
Valdez BC, Li Y, Murray D, Liu Y, Nieto Y, Champlin RE, Andersson BS. The PARP inhibitor olaparib enhances the cytotoxicity of combined gemcitabine, busulfan and melphalan in lymphoma cells. Leuk Lymphoma 2017; 58:2705-2716. [PMID: 28394191 DOI: 10.1080/10428194.2017.1306647] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
The combination of gemcitabine (Gem), busulfan (Bu), and melphalan (Mel) is a promising regimen for autologous stem-cell transplantation (SCT) for lymphomas. To further improve the efficacy of [Gem + Bu + Mel], we added poly(ADP-ribose) polymerase (PARP) inhibitor olaparib (Ola). We hypothesized that Ola would inhibit the repair of damaged DNA caused by [Gem + Bu + Mel]. Exposure of J45.01 and Toledo cell lines to IC10-20 of individual drug inhibited proliferation by 6-16%; [Gem + Bu + Mel] by 20-27%; and [Gem + Bu + Mel + Ola] by 61-67%. The synergistic cytotoxicity of the four-drug combination may be attributed to activation of the DNA-damage response, inhibition of PARP activity and DNA repair, decreased mitochondrial membrane potential, increased production of reactive oxygen species, and activation of the SAPK/JNK stress signaling pathway, all of which may enhance apoptosis. Similar observations were obtained using mononuclear cells isolated from patients with T-cell lymphocytic leukemia. Our results provide a rationale for undertaking clinical trials of this drug combination for lymphoma patients undergoing SCT.
Collapse
Affiliation(s)
- Benigno C Valdez
- a Department of Stem Cell Transplantation and Cellular Therapy , The University of Texas MD Anderson Cancer Center , Houston , TX , USA
| | - Yang Li
- a Department of Stem Cell Transplantation and Cellular Therapy , The University of Texas MD Anderson Cancer Center , Houston , TX , USA
| | - David Murray
- b Department of Experimental Oncology , Cross Cancer Institute , Edmonton , Canada
| | - Yan Liu
- a Department of Stem Cell Transplantation and Cellular Therapy , The University of Texas MD Anderson Cancer Center , Houston , TX , USA
| | - Yago Nieto
- a Department of Stem Cell Transplantation and Cellular Therapy , The University of Texas MD Anderson Cancer Center , Houston , TX , USA
| | - Richard E Champlin
- a Department of Stem Cell Transplantation and Cellular Therapy , The University of Texas MD Anderson Cancer Center , Houston , TX , USA
| | - Borje S Andersson
- a Department of Stem Cell Transplantation and Cellular Therapy , The University of Texas MD Anderson Cancer Center , Houston , TX , USA
| |
Collapse
|
17
|
Kim YI, Park SW, Kwon HS, Yang HS, Cho SY, Kim YJ, Lee HJ. Inhibin-α gene mutations and mRNA levels in human lymphoid and myeloid leukemia cells. Int J Oncol 2017; 50:1403-1412. [PMID: 28260095 DOI: 10.3892/ijo.2017.3895] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2016] [Accepted: 12/02/2016] [Indexed: 11/06/2022] Open
Abstract
The inhibin-α gene was identified as a tumor suppressor gene in the gonads and adrenal glands by functional studies using knockout mice. Methylation of CpG sites within the regulatory regions of tumor suppressor gene is frequently associated with their transcriptional silencing. We investigated epigenetic modifications, changes in loss of heterozygosity (LOH), and mutation of the inhibin-α gene, and regulation of transcriptional expression in response to inhibitors of DNA methylation (5-aza-2'-deoxycytidine, 5-AzaC) in human lymphoid (Jurkat, Molt-4, Raji, and IM-9) and myeloid (HL-60, Kasumi-1, and K562) leukemia cells. The inhibin-α promoter was hypermethylated in lymphoid (Molt-4 and Raji) and myeloid (HL-60 and Kasumi-1) leukemia cells. Inhibin-α gene mutations differed significantly between lymphoid (heterozygote) and myeloid (homozygote) leukemia cells. LOH in the inhibin-α gene was detected in lymphoid and myeloid leukemia cells, with the exception of Jurkat cells. Treatment with 5-AzaC, a demethylating agent, resulted in increased inhibin-α mRNA and protein levels in most of the cell lines. Also, 5-AzaC treatment inhibited cell proliferation and induced apoptosis. Taken together, our results reveal that the inhibin-α gene is transcriptionally silenced in human leukemia cells and that reactivation is suppressed by a demethylating agent. In addition, mutations in, and expression levels of, the inhibin-α gene differed between human lymphoid and myeloid leukemia cells.
Collapse
Affiliation(s)
- Young Il Kim
- Medical Science Research Institute, Kyung Hee University Medical Center, Seoul 02447, Republic of Korea
| | - Seung-Won Park
- Department of Biotechnology, Catholic University of Daegu, Daegu 38430, Republic of Korea
| | - Hye Shin Kwon
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Hyung-Seok Yang
- Department of Laboratory Medicine, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Sun Young Cho
- Department of Laboratory Medicine, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Young Jin Kim
- Department of Laboratory Medicine, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Hee Joo Lee
- Department of Laboratory Medicine, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
| |
Collapse
|
18
|
Ghassemi-Barghi N, Etebari M, Jafarian-Dehkordi A. Protective effect of amifostine on busulfan induced DNA damage in human hepatoma cells. Toxicol Mech Methods 2016; 27:52-57. [DOI: 10.1080/15376516.2016.1243601] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Nasrin Ghassemi-Barghi
- Department of Pharmacology and Toxicology Faculty of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran (the Islamic Republic of)
| | - Mahmoud Etebari
- Department of Pharmacology and Toxicology Faculty of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran (the Islamic Republic of)
| | - Abbas Jafarian-Dehkordi
- Department of Pharmacology and Toxicology Faculty of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran (the Islamic Republic of)
| |
Collapse
|
19
|
Kebriaei P, Bassett R, Lyons G, Valdez B, Ledesma C, Rondon G, Oran B, Ciurea S, Alousi A, Popat U, Patel K, Ahmed S, Olson A, Bashir Q, Shah N, Jones R, Marin D, Rezvani K, Nieto Y, Khouri I, Qazilbash M, Hosing C, Shpall E, Champlin RE, Andersson BS. Clofarabine Plus Busulfan is an Effective Conditioning Regimen for Allogeneic Hematopoietic Stem Cell Transplantation in Patients with Acute Lymphoblastic Leukemia: Long-Term Study Results. Biol Blood Marrow Transplant 2016; 23:285-292. [PMID: 27816651 DOI: 10.1016/j.bbmt.2016.11.001] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2016] [Accepted: 11/01/2016] [Indexed: 11/25/2022]
Abstract
We investigated the long-term safety and disease control data obtained with i.v. busulfan (Bu) combined with clofarabine (Clo) in patients with acute lymphoblastic leukemia (ALL) undergoing allogeneic hematopoietic stem cell transplantation (HSCT). A total of 107 patients, median age 38 years (range, 19 to 64 years) received a matched sibling donor (n = 52) or matched unrelated donor (n = 55) transplant for ALL in first complete remission (n = 62), second complete remission (n = 28), or more advanced disease (n = 17). Nearly one-half of the patients had a high-risk cytogenetic profile as defined by the presence of t(9;22) (n = 34), t(4;11) (n = 4), or complex cytogenetics (n = 7). Clo 40 mg/m2 was given once daily, with each dose followed by pharmacokinetically dosed Bu infused over 3 hours daily for 4 days, followed by hematopoietic cell infusion after 2 days of rest. The Bu dose was based on the drug clearance as determined by a test Bu dose of 32 mg/m2. The target daily area under the curve was 5500 µmol/min for patients aged <60 years and 4000 µmol/min for patients aged >59 years. With a median follow-up of 3.3 years among surviving patients (range, 1 to 5.8 years), the 2-year progression-free survival (PFS) for patients undergoing HSCT in first complete remission (CR1), second complete remission (CR2), or more advanced disease was 62%, 34%, and 35%, respectively. The regimen was well tolerated, with nonrelapse mortality (NRM) of 10% at 100 days and 31% at 2 years post-HSCT. The incidence of grade II-IV and III-IV acute graft-versus-host disease (GVHD) was 35% and 10%, respectively; 18% patients developed extensive chronic GVHD. The 2-year overall survival (OS) for patients undergoing HSCT in CR1, CR2, or more advanced disease was 70%, 57%, and 35%, respectively. Among 11 patients aged >59 years treated with reduced-dose Bu in CR1 (n = 7) or CR2 (n = 4), 4 remain alive and disease-free, with a median follow-up of 2.6 years (range, 2 to 4.7 years). Only the presence of minimal residual disease at the time of transplantation was associated with significantly worse PFS and OS in multivariate analysis. Our data indicate that the Clo-Bu combination provides effective disease control while maintaining a favorable safety profile. OS and NRM rates compare favorably with those for traditional myeloablative total body irradiation-based conditioning regimens.
Collapse
Affiliation(s)
- Partow Kebriaei
- Department of Stem Cell Transplantation and Cellular Therapy, University of Texas M.D. Anderson Cancer Center, Houston, Texas.
| | - Roland Bassett
- Department of Biostatistics, University of Texas M.D. Anderson Cancer Center, Houston, Texas
| | - Genevieve Lyons
- Department of Biostatistics, University of Texas M.D. Anderson Cancer Center, Houston, Texas
| | - Ben Valdez
- Department of Stem Cell Transplantation and Cellular Therapy, University of Texas M.D. Anderson Cancer Center, Houston, Texas
| | - Celina Ledesma
- Department of Stem Cell Transplantation and Cellular Therapy, University of Texas M.D. Anderson Cancer Center, Houston, Texas
| | - Gabriela Rondon
- Department of Stem Cell Transplantation and Cellular Therapy, University of Texas M.D. Anderson Cancer Center, Houston, Texas
| | - Betul Oran
- Department of Stem Cell Transplantation and Cellular Therapy, University of Texas M.D. Anderson Cancer Center, Houston, Texas
| | - Stefan Ciurea
- Department of Stem Cell Transplantation and Cellular Therapy, University of Texas M.D. Anderson Cancer Center, Houston, Texas
| | - Amin Alousi
- Department of Stem Cell Transplantation and Cellular Therapy, University of Texas M.D. Anderson Cancer Center, Houston, Texas
| | - Uday Popat
- Department of Stem Cell Transplantation and Cellular Therapy, University of Texas M.D. Anderson Cancer Center, Houston, Texas
| | - Krina Patel
- Department of Stem Cell Transplantation and Cellular Therapy, University of Texas M.D. Anderson Cancer Center, Houston, Texas
| | - Sairah Ahmed
- Department of Stem Cell Transplantation and Cellular Therapy, University of Texas M.D. Anderson Cancer Center, Houston, Texas
| | - Amanda Olson
- Department of Stem Cell Transplantation and Cellular Therapy, University of Texas M.D. Anderson Cancer Center, Houston, Texas
| | - Qaiser Bashir
- Department of Stem Cell Transplantation and Cellular Therapy, University of Texas M.D. Anderson Cancer Center, Houston, Texas
| | - Nina Shah
- Department of Stem Cell Transplantation and Cellular Therapy, University of Texas M.D. Anderson Cancer Center, Houston, Texas
| | - Roy Jones
- Department of Stem Cell Transplantation and Cellular Therapy, University of Texas M.D. Anderson Cancer Center, Houston, Texas
| | - David Marin
- Department of Stem Cell Transplantation and Cellular Therapy, University of Texas M.D. Anderson Cancer Center, Houston, Texas
| | - Katayoun Rezvani
- Department of Stem Cell Transplantation and Cellular Therapy, University of Texas M.D. Anderson Cancer Center, Houston, Texas
| | - Yago Nieto
- Department of Stem Cell Transplantation and Cellular Therapy, University of Texas M.D. Anderson Cancer Center, Houston, Texas
| | - Issa Khouri
- Department of Stem Cell Transplantation and Cellular Therapy, University of Texas M.D. Anderson Cancer Center, Houston, Texas
| | - Muzaffar Qazilbash
- Department of Stem Cell Transplantation and Cellular Therapy, University of Texas M.D. Anderson Cancer Center, Houston, Texas
| | - Chitra Hosing
- Department of Stem Cell Transplantation and Cellular Therapy, University of Texas M.D. Anderson Cancer Center, Houston, Texas
| | - Elizabeth Shpall
- Department of Stem Cell Transplantation and Cellular Therapy, University of Texas M.D. Anderson Cancer Center, Houston, Texas
| | - Richard E Champlin
- Department of Stem Cell Transplantation and Cellular Therapy, University of Texas M.D. Anderson Cancer Center, Houston, Texas
| | - Borje S Andersson
- Department of Stem Cell Transplantation and Cellular Therapy, University of Texas M.D. Anderson Cancer Center, Houston, Texas
| |
Collapse
|
20
|
Romidepsin targets multiple survival signaling pathways in malignant T cells. Blood Cancer J 2015; 5:e357. [PMID: 26473529 PMCID: PMC4635192 DOI: 10.1038/bcj.2015.83] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2015] [Accepted: 09/02/2015] [Indexed: 01/24/2023] Open
Abstract
Romidepsin is a cyclic molecule that inhibits histone deacetylases. It is Food and Drug Administration-approved for treatment of cutaneous and peripheral T-cell lymphoma, but its precise mechanism of action against malignant T cells is unknown. To better understand the biological effects of romidepsin in these cells, we exposed PEER and SUPT1 T-cell lines, and a primary sample from T-cell lymphoma patient (Patient J) to romidepsin. We then examined the consequences in some key oncogenic signaling pathways. Romidepsin displayed IC50 values of 10.8, 7.9 and 7.0 nm in PEER, SUPT1 and Patient J cells, respectively. Strong inhibition of histone deacetylases and demethylases, increased production of reactive oxygen species and decreased mitochondrial membrane potential were observed, which may contribute to the observed DNA-damage response and apoptosis. The stress-activated protein kinase/c-Jun N-terminal kinase signaling pathway and unfolded protein response in the endoplasmic reticulum were activated, whereas the phosphatidylinositol 3-kinase/AKT/mammalian target of rapamycin (PI3K/AKT/mTOR) and β-catenin pro-survival pathways were inhibited. The decreased level of β-catenin correlated with the upregulation of its inhibitor SFRP1 through romidepsin-mediated hypomethylation of its gene promoter. Our results provide new insights into how romidepsin invokes malignant T-cell killing, show evidence of its associated DNA hypomethylating activity and offer a rationale for the development of romidepsin-containing combination therapies.
Collapse
|
21
|
Valdez BC, Li Y, Murray D, Ji J, Liu Y, Popat U, Champlin RE, Andersson BS. Comparison of the cytotoxicity of cladribine and clofarabine when combined with fludarabine and busulfan in AML cells: Enhancement of cytotoxicity with epigenetic modulators. Exp Hematol 2015; 43:448-61.e2. [PMID: 25704054 DOI: 10.1016/j.exphem.2015.02.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2014] [Revised: 01/20/2015] [Accepted: 02/12/2015] [Indexed: 11/17/2022]
Abstract
Clofarabine (Clo), fludarabine (Flu), and busulfan (Bu) combinations are efficacious in hematopoietic stem cell transplantation for myeloid leukemia. We sought to determine whether the more affordable drug cladribine (Clad) can provide a viable alternative to Clo, with or without panobinostat (Pano) and 5-aza-2'-deoxycytidine (DAC). Both Clad+Flu+Bu and Clo+Flu+Bu combinations showed synergistic cytotoxicity in KBM3/Bu250(6), HL60, and OCI-AML3 cell lines. Cell exposure to these drug combinations resulted in 60%-80% inhibition of proliferation; activation of the ATM pathway; increase in histone modifications; decrease in HDAC3, HDAC4, HDAC5 and SirT7 proteins; decrease in mitochondrial membrane potential; activation of apoptosis and stress signaling pathways; and downregulation of the AKT pathway. These drug combinations activated DNA-damage response and apoptosis in primary cell samples from AML patients. At lower concentrations of Clad/Clo, Flu, and Bu, inclusion of Pano and DAC enhanced cell killing, increased histone modifications and DNA demethylation, and increased the levels of P16/INK4a, P15/INK4b and P21/Waf1/Cip1 proteins. The observed DNA demethylating activity of Clad and Clo may complement DAC activity; increase demethylation of the gene promoters for SFRP1, DKK3, and WIF1; and cause degradation of β-catenin in cells exposed to Clad/Clo+Flu+Bu+DAC+Pano. The overlapping activities of Clad/Clo+Flu+Bu, Pano, and DAC in DNA-damage formation and repair, histone modifications, DNA demethylation, and apoptosis may underlie their synergism. Our results provide a basis for supplanting Clo with Clad and for including epigenetic modifiers in the pre-hematopoietic stem cell transplantation conditioning regimen for myeloid leukemia patients.
Collapse
Affiliation(s)
- Benigno C Valdez
- Department of Stem Cell Transplantation and Cellular Therapy, University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | - Yang Li
- Department of Stem Cell Transplantation and Cellular Therapy, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - David Murray
- Department of Experimental Oncology, Cross Cancer Institute, Edmonton, Alberta, Canada
| | - Jie Ji
- Department of Stem Cell Transplantation and Cellular Therapy, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Yan Liu
- Department of Stem Cell Transplantation and Cellular Therapy, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Uday Popat
- Department of Stem Cell Transplantation and Cellular Therapy, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Richard E Champlin
- Department of Stem Cell Transplantation and Cellular Therapy, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Borje S Andersson
- Department of Stem Cell Transplantation and Cellular Therapy, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
22
|
Tyagi T, Treas JN, Mahalingaiah PKS, Singh KP. Potentiation of growth inhibition and epigenetic modulation by combination of green tea polyphenol and 5-aza-2′-deoxycytidine in human breast cancer cells. Breast Cancer Res Treat 2015; 149:655-68. [DOI: 10.1007/s10549-015-3295-5] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2014] [Accepted: 02/02/2015] [Indexed: 12/18/2022]
|
23
|
Jeong M, Goodell MA. New answers to old questions from genome-wide maps of DNA methylation in hematopoietic cells. Exp Hematol 2014; 42:609-17. [PMID: 24993071 PMCID: PMC4137036 DOI: 10.1016/j.exphem.2014.04.008] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2014] [Revised: 04/17/2014] [Accepted: 04/22/2014] [Indexed: 01/10/2023]
Abstract
DNA methylation is a well-studied epigenetic modification essential for efficient cellular differentiation. Aberrant DNA methylation patterns are a characteristic feature of cancer, including myeloid malignancies such as acute myeloid leukemia. Recurrent mutations in DNA-modifying enzymes were identified in acute myeloid leukemia and linked to distinct DNA methylation signatures. In addition, discovery of Tet enzymes provided new mechanisms for the reversal of DNA methylation. Advances in base-resolution profiling of DNA methylation have enabled a more comprehensive understanding of the methylome landscape in the genome. This review will summarize and discuss the key questions in the function of DNA methylation in the hematopoietic system, including where and how DNA methylation regulates diverse biological processes in the genome as elucidated by recent studies.
Collapse
Affiliation(s)
- Mira Jeong
- Stem Cells and Regenerative Medicine Center, Department of Pediatrics and Molecular & Human Genetics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Margaret A. Goodell
- Stem Cells and Regenerative Medicine Center, Department of Pediatrics and Molecular & Human Genetics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| |
Collapse
|
24
|
Mitomi T, Kawano Y, Kinoshita-Kawano S. Effect of the antineoplastic agent busulfan on rat molar root development. Arch Oral Biol 2014; 59:47-59. [PMID: 24404577 DOI: 10.1016/j.archoralbio.2013.09.009] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
OBJECTIVE The antineoplastic bifunctional-alkylating agent busulfan (Bu) induces developmental anomalies. We examined histopathological changes in the molar roots of rats that received Bu at different stages of root formation. DESIGN At different developmental stages, i.e., on postnatal days (P) 13, 15, and 19, rats were administered 7.5 mg/kg of Bu dissolved in dimethyl sulfoxide (DMSO) and then killed on P 30. After micro-computed tomography analysis, the maxillary first molars underwent immunohistochemical analysis for cytokeratin 14 (CK14), nestin, and dentin sialoprotein (Dsp). This was followed by histomorphometric analysis. RESULTS The rats receiving Bu at an early stage (i.e., P 13 and P 15) showed osteodentin formation and complete destruction of the Hertwig's epithelial root sheath (HERS). Cells around osteodentin showed nestin and Dsp immunoreactivity. The root lengths in rats treated with Bu at P 13 (1228.44 ± 62.17 μm) and P 15 (1536.08 ± 109.71 μm) were lower than that in the control rats (1674.10 ± 40 μm). A narrowed apical foramen and an increased amount of osteodentin were also present, depending on the rat's age at the time of treatment (P < 0.05). CONCLUSION Busulfan treatment in juvenile rats resulted in abnormal root development, depending on the stage at which Bu was administered. This abnormal development may result from the destruction of the HERS. The administration of Bu caused a shortage of HERS cells, which are required for normal root development. This disturbs root formation, resulting in osteodentin formation and a narrowed apex foramen.
Collapse
|
25
|
The interplay between epigenetic silencing, oncogenic KRas and HIF-1 regulatory pathways in control of BNIP3 expression in human colorectal cancer cells. Biochem Biophys Res Commun 2013; 441:707-12. [PMID: 24211581 DOI: 10.1016/j.bbrc.2013.10.098] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2013] [Accepted: 10/18/2013] [Indexed: 01/22/2023]
Abstract
Bcl-2/adenovirus E1B-19kDa-interacting protein 3 (BNIP3) is an important mediator of cell survival and a member of the Bcl-2 family of proteins that regulate programmed cell death and autophagy. We have previously established a link between the expression of oncogenic HRas and up-regulation of BNIP3 and the control of autophagy in cancer cells. However, in view of varied expression of BNIP3 in different tumor types and emerging uncertainties as to the role of epigenetic silencing, oncogenic regulation and the role of BNIP3 in cancer are still poorly understood. In the present study we describe profound effect of KRas on the expression of methylated BNIP3 in colorectal cancer cells and explore the interplay between HIF-1, hypoxia pathway and oncogenic KRas in this context. We observed that BNIP3 mRNA remains undetectable in aggressive DLD-1 cells harboring G13D mutant KRAS and HT-29 colorectal cancer cells unless the cells are exposed to demethylating agents such as 5-aza-2'-deoxycytidine. Following this treatment BNIP3 expression remains uniquely dependent on the Ras activity. We found that hypoxia or pharmacological activation of HIF-1 alone contributes to, but is not sufficient for efficient induction of BNIP3 mRNA transcription in cells lacking mutant KRas activity. The up-regulation of BNIP3 by KRas in this setting is mediated by the MAPK pathway, and is attenuated by the respective inhibitors (PD98059, U0126). Thus, we demonstrate the novel mechanism where activity of Ras is essential for 5-aza-2'-deoxycytidine-mediated BNIP3 expression. Moreover, we found that 5-aza-2'-deoxycytidine-mediated or enforced up-regulation of BNIP3 in DLD-1 cells results in KRas-dependent resistance to 5-Fluorouracil.
Collapse
|
26
|
Smac mimetic and demethylating agents synergistically trigger cell death in acute myeloid leukemia cells and overcome apoptosis resistance by inducing necroptosis. Cell Death Dis 2013; 4:e802. [PMID: 24030154 PMCID: PMC3789178 DOI: 10.1038/cddis.2013.320] [Citation(s) in RCA: 99] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2013] [Revised: 07/25/2013] [Accepted: 07/29/2013] [Indexed: 12/25/2022]
Abstract
Evasion of apoptosis, for example, by inhibitor of apoptosis (IAP) proteins, contributes to treatment resistance and poor outcome in acute myeloid leukemia (AML). Here we identify a novel synergistic interaction between the small-molecule second mitochondria-derived activator of caspases (Smac) mimetic BV6, which antagonizes X-linked IAP, cellular IAP (cIAP)1 and cIAP2, and the demethylating agents 5-azacytidine or 5-aza-2'-deoxycytidine (DAC) to induce cell death in AML cells, including apoptosis-resistant cells. Calculation of combination index (CI) confirms that this drug combination is highly synergistic (CI 0.02-0.4). In contrast, BV6 and DAC at equimolar concentrations do not cause synergistic toxicity against normal peripheral blood lymphocytes, pointing to some tumor cell selectivity. Molecular studies reveal that BV6 and DAC cooperate to trigger the activation of caspases, mitochondrial perturbations and DNA fragmentation, consistent with apoptotic cell death. However, the broad-range caspase inhibitor N-benzyloxycarbonyl-Val-Ala-Asp-fluoromethylketone (zVAD.fmk) fails to protect against BV6/DAC-induced cell death and even significantly increases the percentage of Annexin-V/propidium iodide double-positive cells. Importantly, BV6/DAC-induced cell death in the presence of zVAD.fmk is significantly reduced by pharmacological inhibition of key components of necroptosis signaling, that is, receptor-interacting protein (RIP) 1 using necrostatin-1 or mixed lineage kinase domain-like protein (MLKL) using necrosulfonamide. This indicates a switch from BV6/DAC-induced cell death from apoptosis to necroptosis upon caspase inhibition. Thus, BV6 cooperates with demethylating agents to induce cell death in AML cells and circumvents apoptosis resistance via a switch to necroptosis as an alternative mode of cell death. The identification of a novel synergism of BV6 and demethylating agents has important implications for the development of new treatment strategies for AML.
Collapse
|
27
|
Liu J, Xie YS, Wang FL, Zhang LJ, Zhang Y, Luo HS. Cytotoxicity of 5-Aza-2'-deoxycytidine against gastric cancer involves DNA damage in an ATM-P53 dependent signaling pathway and demethylation of P16(INK4A). Biomed Pharmacother 2013; 67:78-87. [PMID: 23201008 DOI: 10.1016/j.biopha.2012.10.015] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2012] [Accepted: 10/22/2012] [Indexed: 01/06/2023] Open
Abstract
The DNA methylation inhibitor 5-Aza-2'-deoxycytidine (5-Aza-CdR) has increasingly attracted worldwide attention for its antineoplastic potential. The cytotoxitic mechanisms, however, especially, the relative contribution of silenced genes reactivation by demethylation and enzyme-DNA adduct formation to the efficacy of 5-Aza-CdR is still a crucial unresolved question. In this investigation, we demonstrated that 5-Aza-CdR treatment resulted in growth suppression in a concentration and time-dependent manner and G2 phrase arrest - hallmarks of a DNA damage response in gastric cancer AGS cells. Formation of DNA double-strand breaks, as monitored by comet assay was examined in an ATM (ataxia-telangiectasia mutated)-dependent manner based on the fact that PI3K inhibitor Wortmannin abolished the action of cytotoxicity of 5-Aza-CdR. Upon treatment with 5-Aza-CdR, ATM activation was clearly associated with P53 phosphorylation at Ser(15), which was directly responsible for 5-Aza-CdR modified P21(Waf1/Cip1) expression. Further exploration revealed that demethylation of P16(INK4A) correlated with the strikingly down-regulated expressions of DNA methyltransferase 3A as well as 3B was, at least in part, attributed to the cytotoxicity of 5-Aza-CdR in AGS cells. Conclusively, these results greatly enhance our understanding of the mechanisms of cytotoxicity of 5-Aza-CdR and strongly provide the preclinical rationale for an assessment of 5-Aza-CdR to ameliorate patient outcome with gastric cancer.
Collapse
Affiliation(s)
- Juan Liu
- Department of Geriatrics, Renmin Hospital of Wuhan University, Wuchang District, Wuhan, PR China
| | | | | | | | | | | |
Collapse
|
28
|
Ohira T, Ando R, Saito T, Yahata M, Oshima Y, Tamura K. Busulfan-induced pathological changes of the cerebellar development in infant rats. ACTA ACUST UNITED AC 2012; 65:789-97. [PMID: 23276622 DOI: 10.1016/j.etp.2012.11.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2012] [Revised: 10/18/2012] [Accepted: 11/22/2012] [Indexed: 11/17/2022]
Abstract
Busulfan, an antineoplastic bifunctional-alkylating agent, is known to induce developmental anomalies and fetal neurotoxicity. We previously reported that busulfan induced p53-dependent neural progenitor cell apoptosis in fetal rat brain (Ohira et al., 2012). The present study was carried out to clarify the characteristics and sequence of busulfan-induced pathological changes in infant rat brain. Six-day-old male infant rats were treated with 10, 20, 30 or 50 mg/kg of busulfan, and their brains were examined at 1, 2, 4, 7, and 14 days after treatment (DAT). As a result, histopathological changes were selectively detected in the external granular layer (EGL), deep cerebellar nuclei (DCN) and cerebellar white matter (CWM) in the cerebellum with dose-dependent severity but not in the cerebrum. In the normal infant rat cerebellum, granular cells in the EGL were proliferating and moving to the internal granular layer during the normal developmental process. In the EGL of the busulfan group, apoptotic granular cells increased at 2 DAT simultaneously with increased numbers of p53- and p21-positive cells while mitotic granular cells decreased, suggesting an occurrence of p53-related apoptosis and depression of proliferative activity in granular cells. In the DCN, apoptotic glial cells increased at 2 DAT and glial cells showing abnormal mitosis increased at 4 DAT. In the CWN, edematous change accompanying a few apoptotic cells was found in the CWN, especially in the parafolliculus (PFL), from 2 to 7 DAT. The present study demonstrated for the first time the characteristics and sequence of busulfan-induced pathological changes in infant rat cerebellum.
Collapse
Affiliation(s)
- Toko Ohira
- Gotemba Laboratories, Biology and Zoology Research Center Inc., 1284, Kamado, Gotemba, Shizuoka 412-0039, Japan.
| | | | | | | | | | | |
Collapse
|
29
|
Kebriaei P, Basset R, Ledesma C, Ciurea S, Parmar S, Shpall EJ, Hosing C, Khouri I, Qazilbash M, Popat U, Alousi A, Nieto Y, Jones RB, de Lima M, Champlin RE, Andersson BS. Clofarabine combined with busulfan provides excellent disease control in adult patients with acute lymphoblastic leukemia undergoing allogeneic hematopoietic stem cell transplantation. Biol Blood Marrow Transplant 2012; 18:1819-26. [PMID: 22750645 PMCID: PMC4319530 DOI: 10.1016/j.bbmt.2012.06.010] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2012] [Accepted: 06/13/2012] [Indexed: 11/21/2022]
Abstract
We investigated the safety and early disease control data for i.v. busulfan (Bu) in combination with clofarabine (Clo) in patients with acute lymphoblastic leukemia undergoing allogeneic hematopoietic stem cell transplantation (SCT). Fifty-one patients (median age, 36 years; range, 20-64 years) received a matched sibling (n = 24), syngeneic (n = 2), or matched unrelated donor transplant (n = 25) for acute lymphoblastic leukemia in first complete remission (n = 30), second complete remission (n = 13), or active disease (n = 8). More than one-half of the patients had a high-risk cytogenetic profile, as defined by the presence of t(9;22) (n = 17), t(4;11) (n = 3), or complex cytogenetics (n = 7). Clo 40 mg/m(2) was given once daily, with each dose followed by pharmacokinetically dosed Bu infused over 3 hours daily for 4 days, followed by hematopoietic SCT 2 days later. The Bu dose was based on drug clearance, as determined by the patient's response to a 32-mg/m(2) Bu test dose given 48 hours before the high-dose regimen. The target daily area under the receiver-operating characteristic curve was 5500 μM/min for patients age <60 years and 4000 μM/min for those age ≥60 years. The regimen was well tolerated, with a 100-day nonrelapse mortality rate of 6%. With a median follow-up of 14 months among surviving patients (range, 6-28 months), the 1-year overall survival, disease-free survival, and nonrelapse mortality rates were 67% (95% confidence interval [CI], 55%-83%), 54% (95% CI, 41%-71%), and 32% (95% CI, 16%-45%), respectively. For patients undergoing SCT in first remission, these respective rates were 74%, 64%, and 25%. Our data indicate that the combination of Clo and Bu provides effective disease control while maintaining a favorable safety profile.
Collapse
Affiliation(s)
- Partow Kebriaei
- Department of Stem Cell Transplantation and Cellular Therapy, University of Texas M.D. Anderson Cancer Center, Houston, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Zeng J, Wu J, Wang P, Zhang Y. Synergistic effect of 5-aza-2′-deoxycytidine and 5-fluorouracil in human gastric cancer cells. Shijie Huaren Xiaohua Zazhi 2012; 20:3015-3020. [DOI: 10.11569/wcjd.v20.i31.3015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To examine the impact of DNA methyltransferase inhibitor 5-aza-2′-deoxycytidine (5-Aza-CdR) on gastric cancer cells and to unveile the interaction between 5-Aza-CdR and conventional chemotherapeutic agent 5-fluorouracil (5-Fu).
METHODS: Gastric cell lines expressing either wild-type P53 (AGS) or mutant-type P53 (BGC-823) were treated with 2.5 µmol/L of 5-Aza-CdR and/or 5-Fu for 0-96 h. Cell viability and proliferation were determined by 3-(4,5)-dimethylthiahiazo(-z-y1)-3,5-di-phenytetrazoliumromide (MTT) assay. Cell apoptosis was detected by annexin V staining. The activity of caspases was determined, and expression of downstream molecules was detected by Western blot.
RESULTS: Compared to untreated AGS and BGC-823 cells, treatment with 5-Aza-CdR significantly suppressed cell proliferation and viability in both cell lines in a time-dependent manner (both P < 0.01). In addition, 5-Aza-CdR induced marked apoptosis of AGS and BGC-823 cells in a time-dependent manner. However, the stimulation of distinct apoptotic pathways was largely dependent on P53 status. In AGS cells, 5-Aza-CdR-induced apoptosis was mediated by intrinsic apoptotic pathway which was modified dramatically by caspase 9. In BGC-823 cells expressing mutant P53, 5-Aza-CdR-induced apoptosis was dependent on caspase-independent apoptotic signaling due to the fact that we failed to observe elevated caspase activity or expression. 5-Aza-CdR in combination with 5-FU showed significant synergistic effects in both AGS and BGC-823 cells, implying that 5-Aza-CdR could efficaciously sensitize the responses of both cell lines to 5-Fu.
CONCLUSION: Our findings strongly demonstrate that 5-Aza-CdR is a potential anti-gastric cancer candidate. 5-Aza-CdR and 5-Fu have significant synergistic effects in human gastric cancer cells.
Collapse
|
31
|
Tsujioka T, Yokoi A, Uesugi M, Kishimoto M, Tochigi A, Suemori S, Tohyama Y, Tohyama K. Effects of DNA methyltransferase inhibitors (DNMTIs) on MDS-derived cell lines. Exp Hematol 2012; 41:189-97. [PMID: 23085465 DOI: 10.1016/j.exphem.2012.10.006] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2012] [Revised: 10/03/2012] [Accepted: 10/12/2012] [Indexed: 10/27/2022]
Abstract
DNA methyltransferase inhibitors (DNMTIs), including decitabine (DAC) and azacitidine (AZA), have recently been highlighted for the treatment of high-risk myelodysplastic syndrome (MDS); however, their action mechanisms have not been clearly defined. Therefore, we investigated the effects of DNMTIs on MDS-derived cell lines in vitro. An MDS-derived cell line MDS92 and its blastic subline MDS-L and HL-60 were used. All three cell lines were sensitive to DNMTIs, but MDS-L was the most susceptible. DAC-induced cell death in MDS-L was preceded by DNA damage-induced G2 arrest via a p53-independent pathway. AZA did not influence the pattern of cell cycle, although it induced DNA damage response. The IC(50) of DAC or AZA on MDS-L cells was associated with the dose inducing the maximal hypomethylation in long interspersed nuclear elements-1 (LINE-1) methylation assay. AZA suppressed the level of methylation in a time-dependent manner (days 4, 7, and 10), whereas DAC maintained the level of methylation from day 4 to 11. The protein expression of DNMT1 and DNMT3a decreased with the suppression of growth and methylation. We conclude that this study provides in vitro models for understanding the effects of DNMTIs on cell growth and gene regulation, including differences in the possible action mechanism of DAC and AZA.
Collapse
Affiliation(s)
- Takayuki Tsujioka
- Department of Laboratory Medicine, Kawasaki Medical School, 577 Matsushima, Kurashiki, Okayama, Japan.
| | | | | | | | | | | | | | | |
Collapse
|
32
|
Cluzeau T, Robert G, Mounier N, Karsenti JM, Dufies M, Puissant A, Jacquel A, Renneville A, Preudhomme C, Cassuto JP, Raynaud S, Luciano F, Auberger P. BCL2L10 is a predictive factor for resistance to azacitidine in MDS and AML patients. Oncotarget 2012; 3:490-501. [PMID: 22577154 PMCID: PMC3380582 DOI: 10.18632/oncotarget.481] [Citation(s) in RCA: 68] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Azacitidine is the leading compound to treat patients suffering myelodysplastic syndrome (MDS) or AML with less than 30% of blasts, but a majority of patients is primary refractory or rapidly relapses under treatment. These patients have a drastically reduced life expectancy as compared to sensitive patients. Therefore identifying predictive factors for AZA resistance is of great interest to propose alternative therapeutic strategies for non-responsive patients. We generated AZA-resistant myeloid cell line (SKM1-R) that exhibited increased expression of BCL2L10 an anti-apoptotic Bcl-2 member. Importantly, BCL2L10 knockdown sensitized SKM1-R cells to AZA effect suggesting that increased BCL2L10 expression is linked to AZA resistance in SKM1-R. We next established in 77 MDS patients that resistance to AZA is significantly correlated with the percentage of MDS or AML cells expressing BCL2L10. In addition, we showed that the proportion of BCL2L10 positive bone marrow cells can predict overall survival in MDS or AML patients. We propose a convenient assay in which the percentage of BCL2L10 expressing cells as assessed by flow cytometry is predictive of whether or not a patient will become resistant to AZA. Therefore, systematic determination of BCL2L10 expression could be of great interest in newly diagnosed and AZA-treated MDS patients.
Collapse
Affiliation(s)
- Thomas Cluzeau
- INSERM U1065, Centre Mediterranéen de Médecine Moléculaire, Team Cell Death, Differentiation, Inflammation and Cancer, Nice, France
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
SHIN DONGYEOK, PARK YOUSOO, YANG KWANGMO, KIM GIYOUNG, KIM WUNJAE, HAN MINHO, KANG HOSUNG, CHOI YUNGHYUN. Decitabine, a DNA methyltransferase inhibitor, induces apoptosis in human leukemia cells through intracellular reactive oxygen species generation. Int J Oncol 2012; 41:910-8. [DOI: 10.3892/ijo.2012.1546] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2012] [Accepted: 05/16/2012] [Indexed: 11/05/2022] Open
|
34
|
Kim YI, Shim J, Kim BH, Lee SJ, Lee HK, Cho C, Cho BN. Transcriptional silencing of the inhibin-α gene in human gastric carcinoma cells. Int J Oncol 2012; 41:690-700. [PMID: 22581369 DOI: 10.3892/ijo.2012.1472] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2012] [Accepted: 04/06/2012] [Indexed: 11/06/2022] Open
Abstract
Although inhibin was first identified as a hormone regulating pituitary FSH secretion, it was later recognized to act as a tumor suppressor in the gonad and adrenal glands. Recently, the alpha subunit of this dimeric hormone (inhibin‑α) was reported to be involved in prostate tumorigenesis. To identify additional roles outside the reproductive axis, we investigated inhibin‑α gene activity and subsequent cell fate in human gastric cancer cells. The results were as follows: all the gastric cancer cells had at least one of a set of abnormalities including hypermethylation of the promoter, mutation of the 5'‑UTR or allelic imbalance including LOH in the inhibin‑α gene. Hypermethylation of the promoter and mutation of the 5'‑UTR in inhibin‑α were observed in SNU‑1, SNU‑5 and SNU‑484 cells. LOH was observed in AGS, KATO III, SNU‑5, SNU‑484 and SNU‑668 cells. Treatment with 5‑AzaC, a demethylating agent, induced demethylation of the inhibin‑α promoter in the SNU‑1, SNU‑5 and SNU‑484 cell lines, with the CpG5 site being strongly influenced by 5‑AzaC. In addition, inhibin‑α mRNA and protein were maintained at low levels in most of the gastric cancer cell lines. These low levels of mRNA and protein expression could be increased in most lines by treatment with 5‑AzaC. These increased inhibin‑α expression levels seemed to be closely associated with apoptosis and suppression of cell growth. Taken together, our results reveal that the inhibin‑α gene is transcriptionally silenced in human gastric cancer cells, and that reactivation of the gene suppresses their growth characteristics. This suggests that inhibin‑α may have a more general tumor suppressor activity outside the reproductive axis.
Collapse
Affiliation(s)
- Young Il Kim
- Medical Science Research Institute, Kyung Hee University Medical Center, Seoul, Republic of Korea
| | | | | | | | | | | | | |
Collapse
|
35
|
Pettigrew KA, Armstrong RN, Colyer HAA, Zhang SD, Rea IM, Jones RE, Baird DM, Mills KI. Differential TERT promoter methylation and response to 5-aza-2'-deoxycytidine in acute myeloid leukemia cell lines: TERT expression, telomerase activity, telomere length, and cell death. Genes Chromosomes Cancer 2012; 51:768-80. [PMID: 22517724 DOI: 10.1002/gcc.21962] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2011] [Accepted: 03/22/2012] [Indexed: 11/06/2022] Open
Abstract
The catalytic subunit of human telomerase (TERT) is highly expressed in cancer cells, and correlates with complex cytogenetics and disease severity in acute myeloid leukemia (AML). The TERT promoter is situated within a large CpG island, suggesting that expression is methylation-sensitive. Studies suggest a correlation between hypermethylation and TERT overexpression. We investigated the relationship between TERT promoter methylation and expression and telomerase activity in human leukemia and lymphoma cell lines. DAC-induced demethylation and cell death were observed in all three cell lines, as well as telomere shortening in HL-60 cells. DAC treatment reduced TERT expression and telomerase activity in OCI/AML3 and HL-60 cells, but not in U937 cells. Control U937 cells expressed lower levels of TERT mRNA, carried a highly methylated TERT core promoter, and proved more resistant to DAC-induced repression of TERT expression and cell death. AML patients had significantly lower methylation levels at several CpGs than "well elderly" individuals. This study, the first to investigate the relationship between TERT methylation and telomerase activity in leukemia cells, demonstrated a differential methylation pattern and response to DAC in three AML cell lines. We suggest that, although DAC treatment reduces TERT expression and telomerase activity, this is unlikely to occur via direct demethylation of the TERT promoter. However, further investigations on the regions spanning CpGs 7-12 and 14-16 may reveal valuable information regarding transcriptional regulation of TERT.
Collapse
Affiliation(s)
- Kerry A Pettigrew
- Centre for Cancer Research and Cell Biology, Queens University Belfast, Belfast BT9 7BL, UK.
| | | | | | | | | | | | | | | |
Collapse
|
36
|
Sequence of busulfan-induced neural progenitor cell damage in the fetal rat brain. ACTA ACUST UNITED AC 2012; 65:523-30. [PMID: 22494849 DOI: 10.1016/j.etp.2012.03.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2011] [Revised: 02/27/2012] [Accepted: 03/11/2012] [Indexed: 01/12/2023]
Abstract
The sequence of neural progenitor cell (NPC) damage induced in fetal rat brain by transplacental exposure to busulfan, an antineoplastic bifunctional-alkylating agent, on gestational day 13 was examined by immunohistochemical and real-time RT-PCR analyses. Following busulfan treatment, pyknotic NPCs first appeared in the medial layer and then extended to the dorsal layer of the ventricular zone (VZ) of the telencephalon. Pyknotic NPCs that were immunohistochemically positive for cleaved caspase-3, i.e. apoptotic NPCs, began to increase at 24 h after treatment, peaked at 48 h, and returned to the control levels at 96 h. On the other hand, the index (%) of phospho-histone H3-positive NPCs, i.e. mitotic NPCs, and that of BrdU-positive NPCs, i.e. S-phase cells, decreased in accordance with the increase in the index of apoptotic NPCs. Prior to the peak time of apoptotic NPCs, the indices of p53- and p21-positive NPCs peaked at 36 h. In addition, the expression levels of p21 and Puma (p53-target genes) mRNAs were elevated in real-time RT-PCR analysis. These findings indicated that busulfan not only induced apoptosis through the p53-mediated intrinsic pathway but also inhibited cell proliferation in NPCs, resulting in a reduction of the width of the telencephalon. On the other hand, in spite of up-regulation of p21 expression, the expression of cyclin D1, part of the cell cycle machinery of the G1/S transition, and the expression levels of Cdc20 and cyclin B1 which are involved in G2/M transition, showed no changes, giving no possible information of busulfan-induced cell cycle arrest in NPCs.
Collapse
|
37
|
The differentiating and apoptotic effects of 2-aza-5′-deoxycytidine are dependent on the PU.1 expression level in PU.1-transgenic K562 cells. Biochem Biophys Res Commun 2012; 420:775-81. [DOI: 10.1016/j.bbrc.2012.03.071] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2012] [Accepted: 03/13/2012] [Indexed: 01/27/2023]
|
38
|
5-azacytidine and decitabine exert proapoptotic effects on neoplastic mast cells: role of FAS-demethylation and FAS re-expression, and synergism with FAS-ligand. Blood 2012; 119:4242-52. [PMID: 22438247 DOI: 10.1182/blood-2011-09-382770] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Aggressive systemic mastocytosis (ASM) and mast cell leukemia (MCL) are advanced hematopoietic neoplasms with poor prognosis. In these patients, neoplastic mast cells (MCs) are resistant against various drugs. We examined the effects of 2 demethylating agents, 5-azacytidine and decitabine on growth and survival of neoplastic MCs and the MC line HMC-1. Two HMC-1 subclones were used, HMC-1.1 lacking KIT D816V and HMC-1.2 exhibiting KIT D816V. Both agents induced apoptosis in HMC-1.1 and HMC-1.2 cells. Decitabine, but not 5-azacytidine, also produced a G(2)/M cell-cycle arrest in HMC-1 cells. Drug-induced apoptosis was accompanied by cleavage of caspase-8 and caspase-3 as well as FAS-demethylation and FAS-re-expression in neoplastic MCs. Furthermore, both demethylating agents were found to synergize with the FAS-ligand in inducing apoptosis in neoplastic MCs. Correspondingly, siRNA against FAS was found to block drug-induced expression of FAS and drug-induced apoptosis in HMC-1 cells. Neither 5-azacytidine nor decitabine induced substantial apoptosis or growth arrest in normal MCs or normal bone marrow cells. Together, 5-azacytidine and decitabine exert growth-inhibitory and proapoptotic effects in neoplastic MCs. These effects are mediated through "FAS-re-expression" and are augmented by the FAS-ligand. Whether epigenetic drugs produce antineoplastic effects in vivo in patients with ASM and MCL remains to be determined.
Collapse
|
39
|
Ning B, Liu G, Liu Y, Su X, Anderson GJ, Zheng X, Chang Y, Guo M, Liu Y, Zhao Y, Nie G. 5-aza-2'-deoxycytidine activates iron uptake and heme biosynthesis by increasing c-Myc nuclear localization and binding to the E-boxes of transferrin receptor 1 (TfR1) and ferrochelatase (Fech) genes. J Biol Chem 2011; 286:37196-206. [PMID: 21903580 PMCID: PMC3199467 DOI: 10.1074/jbc.m111.258129] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2011] [Revised: 09/05/2011] [Indexed: 12/11/2022] Open
Abstract
The hypomethylating agent 5-aza-2'-deoxycytidine (5-aza-CdR) and its derivatives have been successfully used for the treatment of myelodysplastic syndromes, and they frequently improve the anemia that usually accompanies these disorders. However, the molecular mechanisms underlying this action remain poorly understood. In this study, we used two erythroid models, murine erythroid leukemia cells and erythroid burst-forming unit-derived erythroblasts, to show that 5-aza-CdR induced erythroid differentiation and increased the expression of transferrin receptor 1 (TfR1) and ferrochelatase (Fech), thereby increasing iron uptake and heme biosynthesis. We have identified new regulatory E-boxes that lie outside of CpG islands in the TfR1 and Fech promoters, and the methylation status of these sites can be altered by 5-aza-CdR treatment. This in turn altered the binding of the transcription factor c-Myc to these promoter elements. Furthermore, 5-aza-CdR promoted the nuclear translocation of c-Myc and its binding to Max to form functional complexes. The coordinated actions of 5-aza-CdR on the methylation status of the target genes and in stimulating the nuclear translocation of c-Myc provide new molecular insights into the regulation of E-boxes and explain, at least in part, the increased erythroid response to 5-aza-CdR treatment.
Collapse
Affiliation(s)
- Bo Ning
- From the Chinese Academy of Sciences Key Laboratory for Biological Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology, 11 Zhongguancun Beiyitiao, Beijing 100190, China
- the Department of Chemical Biology and Applied Chemistry, College of Chemistry and Molecular Engineering, Peking University, Beijing 100190, China, and
- the Graduate University of the Chinese Academy of Sciences, Beijing 100049, China
| | - Gang Liu
- From the Chinese Academy of Sciences Key Laboratory for Biological Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology, 11 Zhongguancun Beiyitiao, Beijing 100190, China
- the Graduate University of the Chinese Academy of Sciences, Beijing 100049, China
| | - Yuanyuan Liu
- the First Affiliated Hospital of Jilin University, Changchun, Jilin Province 130021, China
| | - Xiufen Su
- the First Affiliated Hospital of Jilin University, Changchun, Jilin Province 130021, China
| | - Gregory J. Anderson
- the Iron Metabolism Laboratory, Queensland Institute of Medical Research, Brisbane, Queensland 4092, Australia
| | - Xin Zheng
- the Laboratory of Molecular Iron Metabolism, College of Life Science, Hebei Normal University, Shijiazhuang, Hebei Province 050016, China
| | - Yanzhong Chang
- the Laboratory of Molecular Iron Metabolism, College of Life Science, Hebei Normal University, Shijiazhuang, Hebei Province 050016, China
| | - Mingzhou Guo
- the Department of Gastroenterology and Hepatology, Chinese People's Liberation Army General Hospital, Beijing 100853 China
| | - Yuanfang Liu
- the Department of Chemical Biology and Applied Chemistry, College of Chemistry and Molecular Engineering, Peking University, Beijing 100190, China, and
| | - Yuliang Zhao
- From the Chinese Academy of Sciences Key Laboratory for Biological Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology, 11 Zhongguancun Beiyitiao, Beijing 100190, China
| | - Guangjun Nie
- From the Chinese Academy of Sciences Key Laboratory for Biological Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology, 11 Zhongguancun Beiyitiao, Beijing 100190, China
| |
Collapse
|
40
|
Mehndiratta M, Palanichamy JK, Pal A, Bhagat M, Singh A, Sinha S, Chattopadhyay P. CpG hypermethylation of the C-myc promoter by dsRNA results in growth suppression. Mol Pharm 2011; 8:2302-9. [PMID: 21879731 DOI: 10.1021/mp200177z] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Deregulation of the c-myc proto-oncogene plays an important role in carcinogenesis. It is, therefore, commonly found to be overexpressed in various types of tumors. Downregulation of c-myc expression assumes great importance in tumor therapy because of its ability to promote and maintain cancer stem cells. Apart from post-transcriptional gene silencing (PTGS), siRNAs have also been shown to cause transcriptional gene silencing (TGS) through epigenetic modifications of a gene locus. This approach can potentially be used to silence genes for longer periods and at a much lesser dosage than PTGS. In this study, we have examined the effect of transfection of a novel siRNA directed against a CpG island encompassing the CT-I(2) region in the P2 promoter of c-myc in U87MG and other cell lines. Transient transfection with this siRNA resulted in c-myc promoter CpG hypermethylation and decreased expression of c-myc (both mRNA and protein) and its downstream targets. A decrease was also observed in the expression of some stemness markers (oct-4 and nanog). Stable transfection also confirmed the promoter CpG hypermethylation and reduced c-myc expression along with reduced cell proliferation and an increase in apoptosis and senescence. A significant decrease in c-myc levels was also observed in three other cancer cell lines after transient transfection under similar conditions. Thus this novel siRNA has the capability of becoming an effective therapeutic tool in malignancies with overexpression of c-myc and may be of particular use in the eradication of recalcitrant cancer stem cells.
Collapse
Affiliation(s)
- Mohit Mehndiratta
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi 110029, India
| | | | | | | | | | | | | |
Collapse
|
41
|
Kebriaei P, Madden T, Kazerooni R, Wang X, Thall PF, Ledesma C, Nieto Y, Shpall EJ, Hosing C, Qazilbash M, Popat U, Khouri I, Champlin RE, Jones RB, Andersson BS. Intravenous busulfan plus melphalan is a highly effective, well-tolerated preparative regimen for autologous stem cell transplantation in patients with advanced lymphoid malignancies. Biol Blood Marrow Transplant 2010; 17:412-20. [PMID: 20674757 DOI: 10.1016/j.bbmt.2010.07.016] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2010] [Accepted: 07/20/2010] [Indexed: 11/27/2022]
Abstract
We investigated the administration of intravenous (i.v.) busulfan (Bu) combined with melphalan (Mel) in patients with advanced lymphoid malignancies undergoing autologous stem cell transplantation. Bu 130 mg/m(2) was infused daily for 4 days, either as a fixed dose per body surface area (BSA), or to target an average daily area under the curve of 5000 μmol-min, determined by a test dose of i.v. Bu at 32 mg/m(2) given 48 hours prior to the high-dose regimen, followed by a rest day, followed by 2 daily doses of Mel at 70 mg/m(2). Stem cells were infused the following day. Eighty patients had i.v. Bu delivered per test dose guidance. The median daily systemic Bu exposure was 4867 μmol-min. One hundred two patients (Hodgkin lymphoma n = 49, non-Hodgkin lymphoma n = 12, multiple myeloma = 41) with a median age of 44 years (range: 19-65 years) were treated. The 2-year overall survival and progression-free survival rates were 85% and 57%, respectively, for patients with Hodgkin lymphoma, 67% and 64%, respectively, for patients with non-Hodgkin lymphoma, and 82% and 42%, respectively, for patients with multiple myeloma. The regimen was very well tolerated with treatment-related mortality at 100 days, 1 year, and 2 years of 1%, 3%, and 3%, respectively. Intravenous Bu-Mel was well tolerated. Disease control wa encouraging, and should be explored in larger phase II studies.
Collapse
Affiliation(s)
- Partow Kebriaei
- Department of Stem Cell Transplantation and Cellular Therapy, University of Texas M.D. Anderson Cancer Center, Houston, Texas 77030, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Valdez BC, Andersson BS. Interstrand crosslink inducing agents in pretransplant conditioning therapy for hematologic malignancies. ENVIRONMENTAL AND MOLECULAR MUTAGENESIS 2010; 51:659-668. [PMID: 20577993 PMCID: PMC4346159 DOI: 10.1002/em.20603] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2023]
Abstract
Despite successful molecularly targeted, highly specific, therapies for hematologic malignancies, the DNA interstrand crosslinking agents, which are among the oldest and least specific cytotoxic drugs, still have an important role. This is particularly true in stem cell transplantation, where virtually every patient receives conditioning therapy with a DNA-alkylating agent-based program. However, due to concern about serious additive toxicities with combinations of different alkylating drugs, the last several years have seen nucleoside analogs, whose cytotoxic action follows vastly different molecular pathways, introduced in combination with alkylating agents. The mechanistic differences paired with different metabolic pathways for the respective drugs have clinically translated into increased safety without appreciable loss of antileukemic activity. In this report, we review pre-clinical evidence for synergistic antileukemic activity when nucleoside analog(s) and DNA-alkylating agent(s) are combined in the most appropriate manner(s), without a measurable decrease in clinical efficacy compared with the more established alkylating agent combinations. Data from our own laboratory using combinations of fludarabine, clofarabine, and busulfan as prototype representatives for these respective classes of cytotoxic agents are combined with information from other investigators to explain how the observed molecular events will result in greatly enhanced synergistic cytotoxicity. We further present possible mechanistic pathways for such desirable cytotoxic synergism. Finally, we propose how this information-backed hypothesis can be incorporated in the design of the next generation conditioning therapy programs in stem cell transplantation to optimize antileukemic efficacy while still safeguarding patient safety.
Collapse
Affiliation(s)
| | - Borje S. Andersson
- Correspondence to: Borje S. Andersson, Department of Stem Cell Transplantation and Cellular Therapy, UT MD Anderson Cancer Center, Unit 423, 1515 Holcombe Blvd, Houston, TX 77030.
| |
Collapse
|