Oncotarget

Research Papers:

Targeting stemness is an effective strategy to control EML4-ALK+ non-small cell lung cancer cells

Se Jin Oh, Kyung Hee Noh, Young-Ho Lee, Soon-Oh Hong, Kwon-Ho Song, Hyo-Jung Lee, Soyeon Kim, Tae Min Kim, Ju-Hong Jeon, Dong-Wan Kim, Jae Hong Seo and Tae Woo Kim _

PDF  |  HTML  |  Supplementary Files  |  How to cite

Oncotarget. 2015; 6:40255-40267. https://doi.org/10.18632/oncotarget.5434

Metrics: PDF 1786 views  |   HTML 4067 views  |   ?  


Abstract

Se Jin Oh1,2, Kyung Hee Noh1,2, Young-Ho Lee1,2, Soon-Oh Hong1,2,5, Kwon-Ho Song1,2, Hyo-Jung Lee1,2, Soyeon Kim3, Tae Min Kim3, Ju-Hong Jeon4, Jae Hong Seo5, Dong-Wan Kim3, Tae Woo Kim1,2

1Laboratory of Infection and Immunology, Graduate School of Medicine, Korea University, Seoul, Korea

2Department of Biochemistry & Molecular Biology, College of Medicine, Korea University, Seoul, Korea

3Department of Internal Medicine, Seoul National University College of Medicine, Seoul, South Korea

4Department of Physiology, Seoul National University, College of Medicine, Seoul, South Korea

5Division of Oncology, Department of Internal Medicine, College of Medicine, Korea University, Seoul, Republic of Korea

Correspondence to:

Tae Woo Kim, e-mail: [email protected]

Dong-Wan Kim, e-mail: [email protected]

Keywords: EML4-ALK, stemness factor, rapamycin, resistance, NSCLC

Received: July 13, 2015     Accepted: October 09, 2015     Published: October 22, 2015

ABSTRACT

The fusion between anaplastic lymphoma kinase (ALK) and echinoderm microtubule-associated protein-like 4 (EML4) is a causative factor in a unique subset of patients with non-small cell lung carcinoma (NSCLC). Although the inhibitor crizotinib, as it blocks the kinase activity of the resulting EML4-ALK fusion protein, displays remarkable initial responses, a fraction of NSCLC cases eventually become resistant to crizotinib by acquiring mutations in the ALK domain or activating bypass pathways via EGFR, KIT, or KRAS. Cancer stem cell (CSC) theory provides a plausible explanation for acquisition of tumorigenesis and resistance. However, the question as to whether EML4-ALK-driven tumorigenesis is linked with the stem-like property and whether the stemness is an effective target in controlling EML4-ALK+ NSCLC including crizotinib-resistant NSCLC cells has not been addressed. Here, we report that stem-like properties stem from ALK activity in EML4-ALK+ NSCLC cells. Notably, treatment with rapamycin, a CSC targeting agent, attenuates stem-like phenotypes of the EML4-ALK+ cells, which increased capability of tumor formation and higher expression of stemness-associated molecules such as ALDH, NANOG, and OCT4. Importantly, combinational treatment with rapamycin and crizotinib leads to synergistic anti-tumor effects on EML4-ALK+ NSCLC cells as well as on those resistant to crizotinib. Thus, we provide a proof of principle that targeting stemness would be a novel strategy to control intractable EML4-ALK+ NSCLC.


Creative Commons License All site content, except where otherwise noted, is licensed under a Creative Commons Attribution 4.0 License.
PII: 5434