Cancer Res Treat.  2018 Apr;50(2):599-613. 10.4143/crt.2016.357.

Crizotinib in Combination with Everolimus Synergistically Inhibits Proliferation of Anaplastic Lymphoma Kinase‒Positive Anaplastic Large Cell Lymphoma

Affiliations
  • 1Cancer Research Institute, Seoul National University, Seoul, Korea. ssysmc@snu.ac.kr
  • 2Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Korea.
  • 3Department of Internal Medicine, Seoul National University Hospital, Seoul, Korea.
  • 4Biomedical Research Institute, Seoul National University Hospital, Seoul, Korea.

Abstract

PURPOSE
Anaplastic large cell lymphoma (ALCL) is a rare aggresive non-Hodgkin lymphoma, of which over 50% of cases have an aberrant nucleophosmin (NPM)"’anaplastic lymphoma kinase (ALK) fusion protein. Both mechanistic target of rapamycin (mTOR) inhibitor everolimus and ALK inhibitor crizotinib have shown promising antitumor activity in ALK-positive cancer cell lines. However, their combined effect has not yet been investigated.
MATERIALS AND METHODS
We evaluated the anti-proliferative effects of everolimus and/or crizotinib in ALK-positive ALCL cell lines, Karpas 299 and SU-DHL-1, and lung adenocarcinoma cell line, NCI-H2228.
RESULTS
We found that individually, both everolimus and crizotinib potently inhibited cell growth in a dose-dependent manner in both Karpas 299 and SU-DHL-1 cells. A combination of these agents synergistically inhibited proliferation in the two cell lines. Crizotinib down-regulated aberrant AKT and ERK phosphorylation induced by everolimus. Combination treatment also significantly increased G0/G1 cell-cycle arrest, DNA damage, and apoptosis compared with everolimus or crizotinib alone in ALK-positive ALCL cells. In the Karpas 299 xenograft model, the combination treatment exerted a stronger antitumor effect than monotherapies, without significant change in body weight. The synergistic effect of everolimus and crizotinib was also reproduced in the ALK-positive lung adenocarcinoma cell line NCI-H2228. The combination treatment abrogated phosphoinositide 3-kinase/AKT and mTOR signaling pathways with little effect on the Ras/ERK pathway in NCI-H2228 cells.
CONCLUSION
Crizotinib combinedwith everolimus synergistically inhibits proliferation of ALK-positive ALCL cells. Our results suggest that this novel combination is worthy of further clinical development in patients with ALK-positive ALCL.

Keyword

TOR serine-threonine kinases; Crizotinib; Everolimus; Anaplastic large cell lymphoma; Anaplastic lymphoma kinase

MeSH Terms

Adenocarcinoma
Apoptosis
Body Weight
Cell Line
DNA Damage
Everolimus*
Heterografts
Humans
Lung
Lymphoma*
Lymphoma, Large-Cell, Anaplastic*
Lymphoma, Non-Hodgkin
Phosphorylation
Phosphotransferases
Sirolimus
TOR Serine-Threonine Kinases
Everolimus
Phosphotransferases
Sirolimus
TOR Serine-Threonine Kinases

Figure

  • Fig. 1. The cytotoxic effect of everolimus and/or crizotinib on anaplastic lymphoma kinase‒positive anaplastic large cell lymphoma cells. (A) Karpas 299 and SU-DHL-1 cells were treated with increasing concentrations of everolimus or crizotinib for 72 hours. Both everolimus and crizotinib potently inhibited growth in a dose-dependent manner in the two cell lines. (B) Karpas 299 and SU-DHL-1 cells were then incubated with everolimus and/or crizotinib at a fixed ratio of 1:40 for 72 hours. Combination index (CI) values were indicated in the graphs. In all combinations, the CI values are less than 1, indicating synergistic cytotoxic effects of the combination treatment.

  • Fig. 2. The effect of everolimus and/or crizotinib on signaling pathways in anaplastic lymphoma kinase (ALK)‒positive anaplastic large cell lymphoma cells. (A) Karpas 299 and SU-DHL-1 cells were treated with 0-16 nM of everolimus or with 0-80 nM of crizotinib for 24 hours. (B) Karpas 299 and SU-DHL-1 cells were treated with 2 nM everolimus and/or 80 nM crizotinib for 24 hours. The combination treatment more potently inhibited phosphoinositide 3-kinase/AKT, Ras/ERK, and mechanistic target of rapamycin (mTOR) signaling pathways compared with everolimus or crizotinib alone.

  • Fig. 3. Synergistic cytotoxicity of everolimus and crizotinib combination and its effects on signaling pathways in the anaplastic lymphoma kinase (ALK)‒positive lung adenocarcinoma cell line, NCI-H2228. (A) NCI-H2228 cells were treated with everolimus and crizotinib at a fixed ratio of 1:80 for 72 hours. Combination index (CI) values calculated are indicated in the graphs. In all combinations, the CI values were less than 1, indicating synergistic cytotoxic effects of this combination treatment in ALK-positive lung adenocarcinoma cells. (B) To evaluate the effect of everolimus and crizotinib on signaling pathways, NCI-H2228 cells were treated with 0-64 nM of everolimus or with 0-1,280 nM of crizotinib for 24 hours. (C) NCI-H2228 cells were treated with 16 nM everolimus and/or 1,280 nM crizotinib for 24 hours. The combination treatment abrogated phosphorylation of molecules in phosphoinositide 3-kinase/AKT and mechanistic target of rapamycin (mTOR) signaling pathways, while its effects on the Ras/ERK signaling pathway was not apparent.

  • Fig. 4. Cell-cycle arrest and apoptosis induced by everolimus and/or crizotinib in anaplastic lymphoma kinase‒positive anaplastic large cell lymphoma cells. Karpas 299 and SU-DHL-1 cells were treated with 1 nM everolimus and/or 40 nM crizotinib for 72 hours. (A) Cell-cycle distribution was assessed by flow cytometry. The cells were gated to sub-G1, G0/G1, S, and G2 phases according to their DNA contents. The combination of crizotinib and everolimus markedly increased the G0/G1 fraction compared with crizotinib or everolimus alone. **p < 0.01. (B) The cells were stained using Annexin V‒FITC and propidium iodide (PI) for apoptosis analysis by flow cytometry. Annexin V‒positive cells were defined as apoptotic. The data were obtained from three independent experiments. *p < 0.05. (C) Western blot analysis demonstrated that the combination treatment decreased cyclin D1 expression and increased p27 and gamma-H2A.X expression and poly(ADP-ribose) polymerase (PARP) and caspase 3 cleavages.

  • Fig. 5. Effects of everolimus and crizotinib combination on Karpas 299 murine xenografts. Karpas 299 cells were inoculated subcutaneously into mice (n=2 per group). After 3 to 4 weeks, the tumors reached 40 mm3 . Mice were treated with control, everolimus, crizotinib, or combination of both. (A) Tumor volume was monitored weekly from the start of treatment. (B) Body weight after drug administration was monitored weekly. (C) Representative H&E, Ki-67, and terminal deoxynucleotidyl transferase–mediated dUTP nick end (TUNEL) staining in excised tumor of control and everolimus and/or crizotinib treatment group. (D) Western blot analysis revealed that the combination treatment increased poly(ADP-ribose) polymerase (PARP) cleavages in tumor xenograft.

  • Fig. 6. Schematic diagram of signaling pathways in anaplastic lymphoma kinase (ALK)‒positive anaplastic large cell lymphoma (ALCL) cells. (A) Phosphoinositide 3-kinase (PI3K)/AKT and Ras/ERK pathways were activated by the nucleophosmin (NPM)-ALK fusion protein in ALK-positive ALCL cells. (B) Combination treatment of crizotinib and everolimus abrogated the AKT and ERK phosphorylation caused by everolimus.


Reference

References

1. Ferreri AJ, Govi S, Pileri SA, Savage KJ. Anaplastic large cell lymphoma, ALK-positive. Crit Rev Oncol Hematol. 2012; 83:293–302.
Article
2. Savage KJ, Harris NL, Vose JM, Ullrich F, Jaffe ES, Connors JM, et al. ALK- anaplastic large-cell lymphoma is clinically and immunophenotypically different from both ALK+ ALCL and peripheral T-cell lymphoma, not otherwise specified: report from the International Peripheral T-Cell Lymphoma Project. Blood. 2008; 111:5496–504.
Article
3. Marzec M, Kasprzycka M, Liu X, Raghunath PN, Wlodarski P, Wasik MA. Oncogenic tyrosine kinase NPM/ALK induces activation of the MEK/ERK signaling pathway independently of c-Raf. Oncogene. 2007; 26:813–21.
Article
4. Bai RY, Ouyang T, Miething C, Morris SW, Peschel C, Duyster J. Nucleophosmin-anaplastic lymphoma kinase associated with anaplastic large-cell lymphoma activates the phosphatidylinositol 3-kinase/Akt antiapoptotic signaling pathway. Blood. 2000; 96:4319–27.
Article
5. Hamedani FS, Cinar M, Mo Z, Cervania MA, Amin HM, Alkan S. Crizotinib (PF-2341066) induces apoptosis due to downregulation of pSTAT3 and BCL-2 family proteins in NPM-ALK(+) anaplastic large cell lymphoma. Leuk Res. 2014; 38:503–8.
Article
6. Shaw AT, Kim DW, Nakagawa K, Seto T, Crino L, Ahn MJ, et al. Crizotinib versus chemotherapy in advanced ALK-positive lung cancer. N Engl J Med. 2013; 368:2385–94.
Article
7. Huang J, Manning BD. The TSC1-TSC2 complex: a molecular switchboard controlling cell growth. Biochem J. 2008; 412:179–90.
Article
8. Vega F, Medeiros LJ, Leventaki V, Atwell C, Cho-Vega JH, Tian L, et al. Activation of mammalian target of rapamycin signaling pathway contributes to tumor cell survival in anaplastic lymphoma kinase-positive anaplastic large cell lymphoma. Cancer Res. 2006; 66:6589–97.
Article
9. Jundt F, Raetzel N, Muller C, Calkhoven CF, Kley K, Mathas S, et al. A rapamycin derivative (everolimus) controls proliferation through down-regulation of truncated CCAAT enhancer binding protein β and NF-κB activity in Hodgkin and anaplastic large cell lymphomas. Blood. 2005; 106:1801–7.
Article
10. Witzig TE, Reeder CB, LaPlant BR, Gupta M, Johnston PB, Micallef IN, et al. A phase II trial of the oral mTOR inhibitor everolimus in relapsed aggressive lymphoma. Leukemia. 2011; 25:341–7.
Article
11. Schatz JH. Targeting the PI3K/AKT/mTOR pathway in nonHodgkin's lymphoma: results, biology, and development strategies. Curr Oncol Rep. 2011; 13:398–406.
Article
12. Berry T, Luther W, Bhatnagar N, Jamin Y, Poon E, Sanda T, et al. The ALK(F1174L) mutation potentiates the oncogenic activity of MYCN in neuroblastoma. Cancer Cell. 2012; 22:117–30.
Article
13. Copp J, Manning G, Hunter T. TORC-specific phosphorylation of mammalian target of rapamycin (mTOR): phospho-Ser2481 is a marker for intact mTOR signaling complex 2. Cancer Res. 2009; 69:1821–7.
Article
14. Samuels Y, Wang Z, Bardelli A, Silliman N, Ptak J, Szabo S, et al. High frequency of mutations of the PIK3CA gene in human cancers. Science. 2004; 304:554.
15. O'Reilly KE, Rojo F, She QB, Solit D, Mills GB, Smith D, et al. mTOR inhibition induces upstream receptor tyrosine kinase signaling and activates Akt. Cancer Res. 2006; 66:1500–8.
16. Julien LA, Carriere A, Moreau J, Roux PP. mTORC1-activated S6K1 phosphorylates Rictor on threonine 1135 and regulates mTORC2 signaling. Mol Cell Biol. 2010; 30:908–21.
Article
17. Zhang H, Bajraszewski N, Wu E, Wang H, Moseman AP, Dabora SL, et al. PDGFRs are critical for PI3K/Akt activation and negatively regulated by mTOR. J Clin Invest. 2007; 117:730–8.
Article
18. Carracedo A, Ma L, Teruya-Feldstein J, Rojo F, Salmena L, Alimonti A, et al. Inhibition of mTORC1 leads to MAPK pathway activation through a PI3K-dependent feedback loop in human cancer. J Clin Invest. 2008; 118:3065–74.
Article
19. Christensen JG, Zou HY, Arango ME, Li Q, Lee JH, McDonnell SR, et al. Cytoreductive antitumor activity of PF-2341066, a novel inhibitor of anaplastic lymphoma kinase and c-Met, in experimental models of anaplastic large-cell lymphoma. Mol Cancer Ther. 2007; 6(12 Pt 1):3314–22.
Article
20. Gambacorti-Passerini C, Messa C, Pogliani EM. Crizotinib in anaplastic large-cell lymphoma. N Engl J Med. 2011; 364:775–6.
Article
21. Gambacorti Passerini C, Farina F, Stasia A, Redaelli S, Ceccon M, Mologni L, et al. Crizotinib in advanced, chemoresistant anaplastic lymphoma kinase-positive lymphoma patients. J Natl Cancer Inst. 2014; 106:djt378.
Article
22. Megiorni F, McDowell HP, Camero S, Mannarino O, Ceccarelli S, Paiano M, et al. Crizotinib-induced antitumour activity in human alveolar rhabdomyosarcoma cells is not solely dependent on ALK and MET inhibition. J Exp Clin Cancer Res. 2015; 34:112.
Article
23. Xu ZZ, Wang WF, Fu WB, Wang AH, Liu ZY, Chen LY, et al. Combination of rituximab and mammalian target of rapamycin inhibitor everolimus (RAD001) in diffuse large B-cell lymphoma. Leuk Lymphoma. 2014; 55:1151–7.
Article
24. Redaelli S, Ceccon M, Pirola A, Peronaci M, Gambacorti-Passerini C, Mologni L. Synergistic activity of ALK and mTOR inhibitors for the treatment of NPM-ALK positive lymphoma. Blood. 2015; 126:3710.
Article
25. Choo AY, Yoon SO, Kim SG, Roux PP, Blenis J. Rapamycin differentially inhibits S6Ks and 4E-BP1 to mediate cell-type-specific repression of mRNA translation. Proc Natl Acad Sci U S A. 2008; 105:17414–9.
Article
26. Sang J, Acquaviva J, Friedland JC, Smith DL, Sequeira M, Zhang C, et al. Targeted inhibition of the molecular chaperone Hsp90 overcomes ALK inhibitor resistance in non-small cell lung cancer. Cancer Discov. 2013; 3:430–43.
Article
27. Ren H, Chen M, Yue P, Tao H, Owonikoko TK, Ramalingam SS, et al. The combination of RAD001 and NVP-BKM120 synergistically inhibits the growth of lung cancer in vitro and In vivo . Cancer Lett. 2012; 325:139–46.
Article
28. Xu CX, Li Y, Yue P, Owonikoko TK, Ramalingam SS, Khuri FR, et al. The combination of RAD001 and NVP-BEZ235 exerts synergistic anticancer activity against non-small cell lung cancer in vitro and In vivo . PLoS One. 2011; 6:e20899.
Article
29. Marzec M, Kasprzycka M, Liu X, El-Salem M, Halasa K, Raghunath PN, et al. Oncogenic tyrosine kinase NPM/ALK induces activation of the rapamycin-sensitive mTOR signaling pathway. Oncogene. 2007; 26:5606–14.
Article
30. Roskoski R Jr. Anaplastic lymphoma kinase (ALK): structure, oncogenic activation, and pharmacological inhibition. Pharmacol Res. 2013; 68:68–94.
Article
Full Text Links
  • CRT
Actions
Cited
CITED
export Copy
Close
Share
  • Twitter
  • Facebook
Similar articles
Copyright © 2024 by Korean Association of Medical Journal Editors. All rights reserved.     E-mail: koreamed@kamje.or.kr