Please wait a minute...
Frontiers of Medicine

ISSN 2095-0217

ISSN 2095-0225(Online)

CN 11-5983/R

Postal Subscription Code 80-967

2018 Impact Factor: 1.847

Front. Med.    2019, Vol. 13 Issue (3) : 388-397    https://doi.org/10.1007/s11684-018-0624-1
RESEARCH ARTICLE
Homoharringtonine synergy with oridonin in treatment of t(8; 21) acute myeloid leukemia
Weina Zhang, Ying Lu, Tao Zhen, Xinjie Chen, Ming Zhang, Ping Liu, Xiangqin Weng, Bing Chen, Yueying Wang()
State Key Laboratory of Medical Genomics, Shanghai Institute of Hematology, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
 Download: PDF(1208 KB)   HTML
 Export: BibTeX | EndNote | Reference Manager | ProCite | RefWorks
Abstract

Collaboration of c-KIT mutations with AML1–ETO (AE) has been demonstrated to induce t(8; 21) acute myeloid leukemia (AML). Targeted therapies designed to eliminate AE and c-KIT oncoproteins may facilitate effective treatment of t(8; 21) AML. Homoharringtonine (HHT) features activity against tumor cells harboring c-KIT mutations, whereas oridonin can induce t(8; 21) AML cell apoptosis and AE cleavage. Therefore, studies should explore the efficacy of combination therapy with oridonin and HHT in t(8; 21) AML. In this study, we investigated the synergistic effects and mechanism of oridonin combined with HHT in t(8; 21) AML cell line and mouse model. The two drugs synergistically inhibited cell viability and induced significant mitochondrial membrane potential loss and apoptosis. Oridonin and HHT induced significant downregulation of c-KIT and its downstream signaling pathways and promoted AE cleavage. HHT increased intracellular oridonin concentration by modulating the expressions of MRP1 and MDR1, thus enhancing the effects of oridonin. The combination of oridonin and HHT prolonged t(8; 21) leukemia mouse survival. In conclusion, oridonin and HHT exert synergistic effects against t(8; 21) leukemia in vivo and in vitro, thereby indicating that their combination may be an effective therapy for t(8; 21) leukemia.

Keywords AML1–ETO      c-KIT      homoharringtonine      oridonin      t(8      21) AML      synergistic effect     
Corresponding Author(s): Yueying Wang   
Just Accepted Date: 25 July 2018   Online First Date: 13 September 2018    Issue Date: 05 June 2019
 Cite this article:   
Weina Zhang,Ying Lu,Tao Zhen, et al. Homoharringtonine synergy with oridonin in treatment of t(8; 21) acute myeloid leukemia[J]. Front. Med., 2019, 13(3): 388-397.
 URL:  
https://academic.hep.com.cn/fmd/EN/10.1007/s11684-018-0624-1
https://academic.hep.com.cn/fmd/EN/Y2019/V13/I3/388
Fig.1  HHT-induced reduction in CD117 expression and apoptosis in Kasumi-1 cells. (A) Cell viability in Kasumi-1 cells treated with different concentrations of HHT (0.005, 0.01, 0.02, and 0.05 mmol/L) for 48 h. (B) FACS analysis of CD117, CD34, and CD11b surface expression. After HHT treatment (0.01, 0.02, and 0.05 mmol/L) for 48 h, the cells were stained with monoclonal antibodies against CD117 (APC-conjugated), CD34 (PE-Cy7), and CD11b (PE) and then analyzed by flow cytometry. (C) Apoptotic effects of HHT in Kasumi-1 cells. After incubating the cells with 0.01, 0.02, and 0.05 mmol/L HHT, apoptotic cell percentages were assayed by flow cytometry after Annexin V/7-AAD double-staining.
Fig.2  Oridonin synergized with HHT to inhibit Kasumi-1 cell viability. (A) The combined cytotoxic effects of oridonin and HHT were determined by CCK-8 assay. Kasumi-1 cells were treated with different concentrations of oridonin and HHT at a fixed ratio of 150:1 for 48 h. (B) The combined cytotoxic effects of 3.0 mmol/L oridonin and 0.02 mmol/L HHT on Kasumi-1 cells at 24, 48, and 72 h. (C) Synergistic curve of HHT and oridonin. (D) CI values of oridonin and HHT after 48 h incubation at ED75, ED90, and ED95. CI values were calculated using CompuSyn software and expressed as mean±SD from four separate experiments.
Fig.3  Oridonin and HHT induced MMP loss and apoptosis in Kasumi-1 cells. (A) Kasumi-1 cells treated with different concentrations of oridonin and HHT were labeled with DiOC6 and then assayed by flow cytometry. Mean fluorescence of DiOC6, as shown in the histograms, represents MMP. (B) FACS analysis of apoptotic effects of oridonin and HHT. After treatment for 24 h, the cells were subjected to Annexin V/7-AAD double-staining and flow cytometry analysis. (C) Effects of oridonin and HHT on caspase-3, PARP, and Mcl-1 expressions. After treating the cells with the indicated concentrations of oridonin and HHT for 24 h, caspase-3 activation, PARP cleavage, and Mcl-1 downregulations were detected by immunoblotting. The concentration unit “mmol/L” was used for oridonin and HHT treatment (e.g., ori3.0 represents 3.0 mmol/L oridonin).
Fig.4  Expressions of c-KIT oncoprotein and its downstream signaling pathway were downregulated in oridonin+ HHT-treated Kasumi-1 cells. (A) c-KIT expression on cell surface. Mean fluorescence of c-KIT in Kasumi-1 cells exposed to 4.5 mmol/L oridonin, 0.03 mmol/L HHT, or both was analyzed by FACS and was compared with that of untreated control cells. **, P<0.01; ***, P<0.001. (B) Significant c-KIT pathway inhibition was induced by the combination of oridonin and HHT. Phosphorylated and total c-KIT, STAT3, STAT5, ERK, and AKT protein expression levels were evaluated by Western blot analysis using specific antibodies. Equal loading was confirmed with b-actin. These blots are representative of multiple independent experiments. (C) Changes in c-KIT expression after treatment with oridonin and HHT. The chart shows changes in c-KIT mRNA levels, which were normalized to GAPDH levels. Data are mean and SD from three independent experiments performed in triplicate. The concentration unit “mmol/L” was used for oridonin and HHT treatment (e.g., ori3.0 represents 3.0 mmol/L oridonin).
Fig.5  By downregulating MDR1 and MRP1 expressions, HHT increased intracellular oridonin concentrations. (A) Immunoblot analysis of AE status. Changes in AE and ?AE expression in Kasumi-1 cells treated with increasing doses of oridonin, HHT, or both are presented. b-actin was used as internal control. (B) Histograms of FACS analysis of bio-ori. Kasumi-1 cells treated with 50 µmol/L bio-ori with or without 30 nmol/L HHT were fixed, permeabilized, and stained with Sav-APC-Cy7. Fluorescence intensity of Sav was indicative of the bio-ori amount detected by flow cytometry. These results are representative of three independent experiments. (C) MRP1 and MDR1 protein expression levels decreased with increasing dose of HHT. Rat monoclonal antibody to MRP1 and polyclonal MDR1 antibody were used, and b-actin was used as loading control. (D) Cytotoxic effects of oridonin in the presence of specific inhibitors. Oridonin was added to Kasumi-1 cells treated with 50 mmol/L MRP1 inhibitor MK571 or 5 mmol/L MDR1 inhibitor PSC833. After 24 h exposure, the cytotoxic effects of oridonin were measured by CCK-8 assay. FACS analysis of the increases in bio-ori levels (E) and apoptosis (F) facilitated by MK571, PSC833, and HHT. *, P<0.05; **, P<0.01; ***, P<0.001, compared with bio-ori alone. The concentration unit “mmol/L” was used for oridonin and HHT treatment (e.g., ori3.0 represents 3.0 mmol/L oridonin).
Fig.6  Oridonin in combination with HHT exerted distinct antileukemia efficacy in treatment of t(8; 21) AML mouse model. A total of 1 × 105 leukemic cells harboring AE and c-KIT N822K were injected into 8-week-old sublethally irradiated female BALB/c mice. On day 5, the tumor-bearing mice were randomly assigned to groups receiving saline solution (vehicle, n = 7), HHT (0.5 mg/kg per day, n = 7), oridonin (40 mg/kg per day, n = 7), or HHT and oridonin (n = 7). (A) Percentages of GFP-positive cells in the peripheral blood at day 19 were analyzed by FACS. (B) Kaplan–Meier survival curves of leukemic mice treated with vehicle, oridonin, and/or HHT.
1 B Jiao, CF Wu, Y Liang, HM Chen, SM Xiong, B Chen, JY Shi, YY Wang, JH Wang, Y Chen, JM Li, LJ Gu, JY Tang, ZX Shen, BW Gu, WL Zhao, Z Chen, SJ Chen. AML1–ETO9a is correlated with C-KIT overexpression/mutations and indicates poor disease outcome in t(8;21) acute myeloid leukemia-M2. Leukemia 2009; 23(9): 1598–1604
2 LM Kelly, DG Gilliland. Genetics of myeloid leukemias. Annu Rev Genomics Hum Genet 2002; 3(1): 179–198
https://doi.org/10.1146/annurev.genom.3.032802.115046
3 GB Zhou, H Kang, L Wang, L Gao, P Liu, J Xie, FX Zhang, XQ Weng, ZX Shen, J Chen, LJ Gu, M Yan, DE Zhang, SJ Chen, ZY Wang, Z Chen. Oridonin, a diterpenoid extracted from medicinal herbs, targets AML1-ETO fusion protein and shows potent antitumor activity with low adverse effects on t(8;21) leukemia in vitro and in vivo. Blood 2007; 109(8): 3441–3450
https://doi.org/10.1182/blood-2006-06-032250
4 T Zhen, CF Wu, P Liu, HY Wu, GB Zhou, Y Lu, JX Liu, Y Liang, KK Li, YY Wang, YY Xie, MM He, HM Cao, WN Zhang, LM Chen, K Petrie, SJ Chen, Z Chen. Targeting of AML1-ETO in t(8;21) leukemia by oridonin generates a tumor suppressor-like protein. Sci Transl Med 2012; 4(127): 127ra38
https://doi.org/10.1126/scitranslmed.3003562
5 YY Wang, LJ Zhao, CF Wu, P Liu, L Shi, Y Liang, SM Xiong, JQ Mi, Z Chen, R Ren, SJ Chen. C-KIT mutation cooperates with full-length AML1-ETO to induce acute myeloid leukemia in mice. Proc Natl Acad Sci USA 2011; 108(6): 2450–2455
https://doi.org/10.1073/pnas.1019625108
6 YY Wang, GB Zhou, T Yin, B Chen, JY Shi, WX Liang, XL Jin, JH You, G Yang, ZX Shen, J Chen, SM Xiong, GQ Chen, F Xu, YW Liu, Z Chen, SJ Chen. AML1-ETO and C-KIT mutation/overexpression in t(8;21) leukemia: implication in stepwise leukemogenesis and response to Gleevec. Proc Natl Acad Sci USA 2005; 102(4): 1104–1109
https://doi.org/10.1073/pnas.0408831102
7 J Lennartsson, L Ronnstrand. The stem cell factor receptor/c-Kit as a drug target in cancer. Curr Cancer Drug Targets 2006; 6(1): 65–75
https://doi.org/10.2174/156800906775471725
8 HM Kantarjian, M Talpaz, V Santini, A Murgo, B Cheson, SM O’Brien. Homoharringtonine: history, current research, and future direction. Cancer 2001; 92(6): 1591–1605
https://doi.org/10.1002/1097-0142(20010915)92:6<1591::AID-CNCR1485>3.0.CO;2-U
9 F Alvandi, VE Kwitkowski, CW Ko, MD Rothmann, S Ricci, H Saber, D Ghosh, J Brown, E Pfeiler, E Chikhale, J Grillo, J Bullock, R Kane, E Kaminskas, AT Farrell, RUS Pazdur. Food and Drug Administration approval summary: omacetaxine mepesuccinate as treatment for chronic myeloid leukemia. Oncologist 2014; 19(1): 94–99
https://doi.org/10.1634/theoncologist.2013-0077
10 J Jin, JX Wang, FF Chen, DP Wu, J Hu, JF Zhou, JD Hu, JM Wang, JY Li, XJ Huang, J Ma, CY Ji, XP Xu, K Yu, HY Ren, YH Zhou, Y Tong, YJ Lou, WM Ni, HY Tong, HF Wang, YC Mi, X Du, BA Chen, Y Shen, Z Chen, SJ Chen. Homoharringtonine-based induction regimens for patients with de-novo acute myeloid leukaemia: a multicentre, open-label, randomised, controlled phase 3 trial. Lancet Oncol 2013; 14(7): 599–608
https://doi.org/10.1016/S1470-2045(13)70152-9
11 HH Zhu, H Jiang, Q Jiang, JS Jia, YZ Qin, XJ Huang. Homoharringtonine, aclarubicin and cytarabine (HAA) regimen as the first course of induction therapy is highly effective for acute myeloid leukemia with t (8;21). Leuk Res 2016; 44: 40–44
https://doi.org/10.1016/j.leukres.2016.02.012
12 SS Lam, ES Ho, BL He, WW Wong, CY Cher, NK Ng, CH Man, H Gill, AM Cheung, HW Ip, CC So, J Tamburini, CW So, DN Ho, CH Au, TL Chan, ES Ma, R Liang, YL Kwong, AY Leung. Homoharringtonine (omacetaxine mepesuccinate) as an adjunct for FLT3-ITD acute myeloid leukemia. Sci Transl Med 2016; 8(359): 359ra129
https://doi.org/10.1126/scitranslmed.aaf3735
13 Y Jin, Z Lu, K Cao, Y Zhu, Q Chen, F Zhu, C Qian, J Pan. The antitumor activity of homoharringtonine against human mast cells harboring the KIT D816V mutation. Mol Cancer Ther 2010; 9(1): 211–223
https://doi.org/10.1158/1535-7163.MCT-09-0468
14 TC Chou. Theoretical basis, experimental design, and computerized simulation of synergism and antagonism in drug combination studies. Pharmacol Rev 2006; 58(3): 621–681
https://doi.org/10.1124/pr.58.3.10
15 R Tang, AM Faussat, P Majdak, C Marzac, S Dubrulle, Z Marjanovic, O Legrand, JP Marie. Semisynthetic homoharringtonine induces apoptosis via inhibition of protein synthesis and triggers rapid myeloid cell leukemia-1 down-regulation in myeloid leukemia cells. Mol Cancer Ther 2006; 5(3): 723–731
https://doi.org/10.1158/1535-7163.MCT-05-0164
16 JI Fletcher, M Haber, MJ Henderson, MD Norris. ABC transporters in cancer: more than just drug efflux pumps. Nat Rev Cancer 2010; 10(2): 147–156
https://doi.org/10.1038/nrc2789
17 D Koley, AJ Bard. Inhibition of the MRP1-mediated transport of the menadione-glutathione conjugate (thiodione) in HeLa cells as studied by SECM. Proc Natl Acad Sci USA 2012; 109(29): 11522–11527
https://doi.org/10.1073/pnas.1201555109
18 XF Hu, A Slater, DM Wall, JD Parkin, P Kantharidis, JR Zalcberg. Cyclosporin A and PSC 833 prevent up-regulation of MDR1 expression by anthracyclines in a human multidrug-resistant cell line. Clin Cancer Res 1996; 2(4): 713–720
19 W Obuchi, S Ohtsuki, Y Uchida, K Ohmine, T Yamori, T Terasaki. Identification of transporters associated with Etoposide sensitivity of stomach cancer cell lines and methotrexate sensitivity of breast cancer cell lines by quantitative targeted absolute proteomics. Mol Pharmacol 2013; 83(2): 490–500
https://doi.org/10.1124/mol.112.081083
20 M Abbaspour Babaei, B Kamalidehghan, M Saleem, HZ Huri, F Ahmadipour. Receptor tyrosine kinase (c-Kit) inhibitors: a potential therapeutic target in cancer cells. Drug Des Devel Ther 2016; 10: 2443–2459
https://doi.org/10.2147/DDDT.S89114
21 K Stankov, S Popovic, M Mikov. C-KIT signaling in cancer treatment. Curr Pharm Des 2014; 20(17): 2849–2880
https://doi.org/10.2174/13816128113199990593
22 EK Allan, TL Holyoake, AR Craig, HG Jorgensen. Omacetaxine may have a role in chronic myeloid leukaemia eradication through downregulation of Mcl-1 and induction of apoptosis in stem/progenitor cells. Leukemia: official journal of the Leukemia Society of America. Leukemia 2011; 25(6): 985–994
23 R Chen, L Guo, Y Chen, Y Jiang, WG Wierda, W Plunkett. Homoharringtonine reduced Mcl-1 expression and induced apoptosis in chronic lymphocytic leukemia. Blood 2011; 117(1): 156–164
https://doi.org/10.1182/blood-2010-01-262808
[1] Xiaoxiao Chen, Yanjing Tang, Jing Chen, Ru Chen, Longjun Gu, Huiliang Xue, Ci Pan, Jingyan Tang, Shuhong Shen. Homoharringtonine is a safe and effective substitute for anthracyclines in children younger than 2 years old with acute myeloid leukemia[J]. Front. Med., 2019, 13(3): 378-387.
[2] Megan A. Hatlen, Lan Wang, Stephen D. Nimer. AML1-ETO driven acute leukemia: insights into pathogenesis and potential therapeutic approaches[J]. Front Med, 2012, 6(3): 248-262.
[3] LIANG Yumei, LI Xianghong, LU Youyong, LV Yali, ZHONG Mei, PU Xiaolu, LI Wenmei. Prognostic significance of clinicopathologic parameters in gastrointestinal stromal tumors: a study of 156 cases[J]. Front. Med., 2008, 2(1): 87-94.
Viewed
Full text


Abstract

Cited

  Shared   
  Discussed