02.12.2016 Views

Journal Thoracic Oncology

WCLC2016-Abstract-Book_vF-WEB_revNov17-1

WCLC2016-Abstract-Book_vF-WEB_revNov17-1

SHOW MORE
SHOW LESS

You also want an ePaper? Increase the reach of your titles

YUMPU automatically turns print PDFs into web optimized ePapers that Google loves.

Abstracts <strong>Journal</strong> of <strong>Thoracic</strong> <strong>Oncology</strong> • Volume 12 Issue S1 January 2017<br />

POSTER SESSION 3 – P3.01: BIOLOGY/PATHOLOGY<br />

MODELS OF LUNG CANCER –<br />

WEDNESDAY, DECEMBER 7, 2016<br />

P3.01-054 ANTAGONISM OF A NOVEL KINASE, MAP3K19, BY<br />

SPECIFIC SMALL MOLECULE INHIBITORS BLOCKS HUMAN<br />

PRIMARY NSCLC TUMOR GROWTH IN MURINE XENOGRAFT<br />

MODELS<br />

Stefen Boehme, John Ludka, Danielle Ditirro, Tai Wei Ly, Kevin Bacon<br />

Biology, Axikin Pharmaceuticals, Inc., San Diego/CA/United States of America<br />

Background: We have identified a novel serine/threonine protein kinase,<br />

MAP3K19, whose expression in normal lung was predominantly localized<br />

to alveolar and interstitial macrophages, bronchial epithelial cells and<br />

type II pneumocytes of the epithelium. We have also found MAP3K19 to be<br />

expressed in multiple, primary NSCLC tumor samples, as well as human lung<br />

cancer cell lines, including A549. The kinase is transcriptionally upregulated<br />

in cells upon various types of cell stress, including oxidative, endoplasmic<br />

reticulum and osmotic stress. Methods: Using two different murine xenograft<br />

models, we assayed the role of MAP3K19 to inhibit the growth of either<br />

primary human NSCLC tumors and A549 cells using small molecule inhibitors.<br />

Results: The ability of i.v. injected A549 cells to colonize and grow in the lung<br />

was significantly reduced in mice that received orally administered, selective<br />

MAP3K19 inhibitors. Similar results were observed in a subcutaneous<br />

xenograft model, as A549 tumor cell growth was inhibited by both MAP3K19<br />

antagonists and other standard of care kinase inhibitors. These studies also<br />

showed an additive anti-proliferative effect when gefitinib or sorafenib<br />

and MAP3K19 inhibitors were co-administered. Importantly, xenograft<br />

models using primary human NSCLC tumors implanted subcutaneously in<br />

immunodeficient mice showed a statistically significant inhibition of tumor<br />

growth when the mice were treated with the orally administered MAP3K19<br />

antagonists. IHC analysis of the tumors showed that mice treated with<br />

the MAP3K19 inhibitors also had decreased levels of Ki-67, c-myc, p27 and<br />

phospho-Bim staining and increased caspase-3 staining. Conclusion: These<br />

results suggest a molecular mechanism by which MAP3K19 may inhibit tumor<br />

cell growth, and further suggest that inhibition of MAP3K19 either by itself<br />

or in combination with other therapies may represent a new avenue for the<br />

treatment of NSCLC. The clinical development of the MAP3K19 inhibitor is<br />

expected to initiate Phase I clinical trials in early 2017.<br />

Keywords: NSCLC, MAP3K19, novel kinase<br />

POSTER SESSION 3 – P3.01: BIOLOGY/PATHOLOGY<br />

Stem Cells in Lung Cancer –<br />

WEDNESDAY, DECEMBER 7, 2016<br />

P3.01-055 IN VITRO CONSTRUCTION OF LUNG CANCER ORGANOIDS<br />

FROM INDUCED LUNG CANCER STEM LIKE CELLS<br />

Hiroyuki Ogawa 1 , Takashi Aoi 2 , Nahoko Shimizu 1 , Daisuke Hokka 1 , Yugo<br />

Tanaka 1 , Michiyo Koyanagi-Aoi 2 , Yoshimasa Maniwa 1<br />

1 <strong>Thoracic</strong> Surgery, Kobe University Hospital, Kobe City/Japan, 2 Department of Ips<br />

Cell Applicationse, Kobe University Hospital, Kobe City/Japan<br />

Background: Lung cancer stem cells are considered to be responsible for lung<br />

cancer progression. However, little is known about how they actually promote<br />

lung cancer progression and metastasis. Methods: We retrovirally introduced<br />

three defined factors (OCT3/4, SOX2, and KLF4) into lung cancer cell line,<br />

A549. We evaluated cancer stem cell properties in the A549 cells transduced<br />

with the three factors (OSK-A549) in terms of their chemo resistance, and<br />

sphere formation ability. We also assessed lung cancer organoid constructing<br />

ability by co-culturing with mesenchymal stem cells (MSC) and human<br />

umbilical vein epithelial cells (HUVEC). Results: OSK-A549 cells formed<br />

dome-shaped colonies in 10 to 15 days after transfection. These colonies were<br />

picked up for further expansion in DMEM/10%FBS medium, and we named<br />

these cells OSK-A549-Colony cells. Induced OSK-A549-colony cells were more<br />

resistant to cisplatin than parental A549 cells. Cell cycle analysis revealed that<br />

the rate of G0/G1 cells was significantly increased in OSK-A549-colony cells.<br />

Sphere forming ability was enhanced in OSK-A549-colony cells. These results<br />

suggested that OSK-A549-colony cells acquired the properties of lung cancer<br />

stem like cells. Co-culture with MSC and HUVEC showed that A549 and OSK-<br />

A549-colony cells could form large spheres equally, however, HE staining of<br />

spheres revealed that OSK-A549-colony cells could form much denser spheres<br />

than those of parental cells (Figure). As the morphology was similar to real<br />

lung cancer tissue, we named this spheres “lung cancer organoids”.<br />

Conclusion: By introducing defined factors, A549 cells acquired lung cancer<br />

stem cell like properties, and these cells could form lung cancer organoids by<br />

co-culturing with MSC and HUVEC. Analysis of these organoids might enable<br />

us to elucidate the molecular mechanism of lung cancer progression and<br />

metastasis.<br />

Keywords: cancer organoid, lung cancer, Cancer stem cell<br />

POSTER SESSION 3 – P3.01: BIOLOGY/PATHOLOGY<br />

STEM CELLS IN LUNG CANCER –<br />

WEDNESDAY, DECEMBER 7, 2016<br />

P3.01-056 ASSOCIATION OF ANGIOGENESIS, EMT AND STEM CELL<br />

CHARACTERISTICS USING HYPOXIC STRESS IN LUNG CANCER<br />

Chankwon Park 1 , Nahyeon Kang 2 , Hyonsoo Joo 1 , Young Kyoon Kim 1 , Seung<br />

Joon Kim 1<br />

1 Department of Internal Medicine, The Catholic University of Korea, Seoul/Korea,<br />

Republic of, 2 The Cancer Research Institute, College of Medicine, the Catholic<br />

University of Korea, Seoul/Korea, Republic of<br />

Background: Hypoxia, a major phenomenon in solid tumors, can promote the<br />

metastatic potential of tumor cells which is associated with chemoresistance<br />

and poor prognosis. It was reported that various angiogenesis factors<br />

including VEGF and HIF, were associated in cancer development and<br />

progression by hypoxia. In addition, both epithelial-mesenchymal transition<br />

(EMT) and cancer stem cells play an important role in malignant progression<br />

in many human tumors. We investigated the effect of hypoxic stress on<br />

the angiogenesis, EMT and stemness acquisition in lung cancer. Methods:<br />

Normal lung cell (BEAS-2B) and lung cancer cell lines (A549, H292, H226<br />

and H460) were incubated in either normoxic or hypoxic (below 1% O 2<br />

)<br />

conditions. For transcriptome analysis, mRNA of BEAS-2B and A549 cell lines<br />

were analyzed using next-generation sequencing (HiSeq 2500 system). For<br />

further validation, angiogenesis markers were analyzed by western blotting.<br />

EMT was assessed with western blotting, wound healing assay and Matrigel<br />

invasion assay, and stem cell characteristics were assessed with RT-PCR,<br />

immunostaining, soft agar colony formation assay, sphere formation assay<br />

and in vivo mice tumor model. Results: Next-generation sequencing revealed<br />

significant changes in the expression of angiogenesis, EMT and stem cell<br />

markers after hypoxic stress. Among the angiogenesis markers, VEGF and<br />

HIF-2α were increased. EMT markers related in hypoxia showed decrease in<br />

E-cadherin and increase in fibronectin, vimentin, N-cadherin, α-SMA, Snail,<br />

Slug, ZEB1 and ZEB2. Stem cell markers such as CXCR4, Oct4 and Nanog were<br />

increased at least one lung cancer cell line in hypoxic condition compared with<br />

in normoxic condition. Functional assays for EMT and stemness acquisition<br />

indicated that hypoxic stress increased wound healing, Matrigel invasion,<br />

sphere formation and in vivo mice tumor formation. Conclusion: These results<br />

suggest that hypoxia induces angiogenesis markers expression which is<br />

associated with EMT and stemness acquisition in lung cancer.<br />

Keywords: hypoxic stress, angiogenesis, lung cancer<br />

POSTER SESSION 3 – P3.01: BIOLOGY/PATHOLOGY<br />

STEM CELLS IN LUNG CANCER –<br />

WEDNESDAY, DECEMBER 7, 2016<br />

P3.01-057 TGF-Β INDUCED EMT AND STEMNESS CHARACTERISTICS<br />

IN LUNG CANCER<br />

Seung Joon Kim 1 , Nahyeon Kang 2 , Hyonsoo Joo 1 , Chankwon Park 1 , Young<br />

Kyoon Kim 1<br />

1 Department of Internal Medicine, The Catholic University of Korea, Seoul/Korea,<br />

Republic of, 2 The Cancer Research Institute, College of Medicine, the Catholic<br />

University of Korea, Seoul/Korea, Republic of<br />

Copyright © 2016 by the International Association for the Study of Lung Cancer<br />

S605

Hooray! Your file is uploaded and ready to be published.

Saved successfully!

Ooh no, something went wrong!