13.10.2013 Views

Summer Undergraduate Research Program - Fred Hutchinson ...

Summer Undergraduate Research Program - Fred Hutchinson ...

Summer Undergraduate Research Program - Fred Hutchinson ...

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.

Investigating a Small Molecule Inhibitor of Aurora<br />

Kinase-A in Ras-driven Pancreas Cancer Cells<br />

Brian W. Wakefield1,2 ; Amy E. Chang2 ; Vikas K. Goel, Ph.D2 ; Sunil R. Hingorani, M.D., Ph.D. 2<br />

1Whitman College, Walla Walla, WA; 2<strong>Fred</strong> <strong>Hutchinson</strong> Cancer <strong>Research</strong> Center, Seattle, WA<br />

2010 Best Poster Presentation Award<br />

RalGTPase Pulldown Assay: RalA activity decreases with AKAi-1.<br />

Results<br />

Cell Proliferation: AKAi-1 inhibits cell growth in KPC cells.<br />

AKA<br />

P + P+<br />

Fig. 10: Western Blot. KPC cells<br />

treated with AKAi-1 or vehicle for 36<br />

hours. Signal intensity normalized to<br />

GST loading control. Inhibition of<br />

AKA by AKAi-1 causes downregulation<br />

of active RalA.<br />

P + P+<br />

AKA<br />

Conclusions<br />

AKAi-1 treatment in primary KPC cell lines:<br />

Inhibits cell proliferation with increased G2/M arrest<br />

Increases apoptosis<br />

These effects occurred in a concentration-dependent manner.<br />

Abstract<br />

Pancreatic ductal adenocarcinoma (PDA) is the fourth leading cause of cancerrelated<br />

deaths in the United States and, with a five-year survival rate of less<br />

than 5%, is almost uniformly fatal. Current therapies are only able to improve<br />

survival by several weeks and new approaches to the study of this disease are<br />

therefore imperative. Over 90% of pancreas cancers have an activating<br />

mutation in the KRAS proto-oncogene while 75% have an inactivating<br />

mutation in the tumor suppressor gene, TP53. We have shown that<br />

endogenous expression of KrasG12D and Trp53R172H in the pancreata of mice<br />

induces disease that mimics human pancreas cancer from precursor lesions to<br />

fully invasive and metastatic PDA. Direct inhibition of oncogenic KRAS in<br />

patients has proved challenging, causing a shift in focus toward its downstream<br />

effectors. The small GTPase, RalA, is downstream target of Ras signaling,<br />

necessary for Ras-mediated oncogenesis, and is also activated by Aurora<br />

Kinase A (AKA). We describe the effects of a small molecule inhibitor of<br />

AKA (AKAi-1) on primary tumor cells derived from the pancreata of mice<br />

expressing oncogenic KrasG12D and mutant Trp53R172H . Standard assay<br />

methods for apoptosis, proliferation, and cell cycle progression were used.<br />

These experiments were designed to determine whether this drug should<br />

proceed to preclinical trials in the autochthonous mouse model.<br />

CDC25<br />

No additive or synergistic effects were observed with simultaneous treatment<br />

of AKAi-1 and Gemcitabine.<br />

Fig. 3: Aurora Kinase A and Kras converge to activate the small GTPase RalA<br />

These data suggest that AKAi-1 may be effective in treating Ras-driven<br />

carcinomas and justify testing this inhibitor in our preclinical mouse model.<br />

AKAi-1<br />

1 2 5 10 - - - 1 2 5 10 1 2 5 10<br />

(µM)<br />

GEM<br />

- - - - 1 5 10 1 1 1 1 5 5 5 5<br />

(nM)<br />

Fig. 5: Growth inhibition by AKAi-1 in KPC cells. Concentration-dependent inhibition was seen in<br />

cells treated with AKAi-1 as compared to DMSO control. No additive or synergistic effects were<br />

observed from co-administration of AKAi-1 with Gemcitabine.<br />

Cell Cycle: AKAi-1 promotes G2/M arrest in KPC cells<br />

Vehicle<br />

Cell Cycle<br />

Progression<br />

Background<br />

Targeted expression of oncogenic KrasG12D to the pancreas initiates<br />

pancreas cancer1 Concomitant expression of mutant Trp53 cooperates to accelerate widely<br />

metastatic disease2 Future Directions<br />

157<br />

To gain a better understanding of the molecular interactions between Kras<br />

and RalA, we will quantify the effects on RalA activity of silencing KrasG12D using shRNA in an inducible system.<br />

Mouse PDA Cells NIH3T3 Cells<br />

Control V33 V28 V24 V14 C. V28 V24 V14<br />

DOX - + - + - + - + - + + + + +<br />

AKAi-1<br />

0.5 µM<br />

AKAi-1<br />

2.5 µM<br />

AKAi-1<br />

- 0.5 1 2.5 5 - - - 0.5 0.5 1 1<br />

(µM)<br />

GEM<br />

- - - - - - 2 5 2 5 2 5<br />

(nM)<br />

Fig. 6: Effect of AKAi-1on cell cycle progression in KPC cells. Concentration-dependent increase in<br />

G2/M arrest was seen in AKAi-1-treated cells. No additive or synergistic effects were observed from coadministration<br />

of AKAi-1 with Gemcitabine. Fig7: Representative plots.<br />

Apoptosis: AKAi-1 increases apoptosis in KPC cells<br />

Materials and Methods<br />

Experiments were conducted in primary cell lines derived from murine<br />

pancreatic tumors (KPC):<br />

KPC<br />

LSL-KrasG12D activating mutation<br />

LSL-Trp53R172H mutation<br />

Fig. 1: Conditional LSL-KRASG12D and LSL-Trp53R172H (KPC) alleles and generation<br />

of functional mutant via Cre recombinase2 Pdx-1-Cre<br />

Fig. 11: Western blot. KPC cells (left) infected with a Doxycyclin-inducible TripZ vector (V) carrying<br />

KrasG12D shRNA show silencing of oncogenic Kras when exposed to Doxycyclin. NIH3T3 cells (right)<br />

Fig. 2b: Specimen<br />

from same mouse<br />

showing hepatic<br />

metastases (black<br />

arrows) and a large<br />

head-of-the-pancreas<br />

primary tumor ( * ) 2<br />

Fig. 2a: Gross<br />

pathological<br />

photograph of<br />

metastatic<br />

PDA in a KPC<br />

mouse2 expressing wild-type Kras show no effect.<br />

Ras activity will be quantified with a RasGTPase pull-down assay<br />

RalA activity will be quantified with a RalGTPase pull-down assay<br />

We will administer AKAi-1 to KPC animals with established tumors alone<br />

and in combination with Gemcitabine. The results of the in vivo study together<br />

with the in vitro data will help determine whether this drug should proceed to<br />

clinical trials.<br />

Vehicle Control<br />

AKAi-1 0.5µM AKAi-1 1.0µM<br />

Fig. 8: Representative apoptosis flow cytometry data from KPC cells. Double positive (upper right,<br />

circled) population represents late apoptotic and dead cells. Annexin V positive population (lower<br />

right) represents early apoptotic cells.<br />

Fig. 4: Western blot. KPC tumor cells show high levels of AKA expression and expression of<br />

RalA<br />

Fig. 2d: Welldifferentiated<br />

liver<br />

metastasis (m) and<br />

surrounding normal<br />

tissue (h) 2<br />

Fig. 2c: Welldifferentiated,<br />

glandular<br />

PDA2 Cell Proliferation: cells were plated in 96-well plates and exposed to a<br />

spectrum of AKAi-1 and Gemcitabine concentrations alone and in<br />

combination. Proliferation was analyzed by WST-1 and absorbance was<br />

read at 450nm.<br />

References<br />

1.Hingorani, Sunil et al. “Preinvasive and Invasive Ductal Pancreatic Cancer and its Early Detection in the Mouse.”<br />

Cancer Cell 4(2003): 437-450.<br />

2.Hingorani, Sunil et al. “Trp53R172H and KrasG12D cooperate to promote chromosomal instability and widely<br />

metastatic pancreatic ductal adenocarcinoma in mice.” Cancer Cell 7(2005): 469-483<br />

3.Brunner, Thomas et al. “Farnesyltransferase Inhibitors: An Overview of the Results of Preclinical and Clinical<br />

Investigations.” Cancer <strong>Research</strong> 63(2003): 5656-5668.<br />

4.Lim, Kian-Huat et al. “Aurora-A Phosphorylates, Activates, and Relocalizes the Small GTPase RalA.” Molecular<br />

and Cellular Biology 30(2010): 508-523<br />

Cell Cycle and Apoptosis: cells were plated in 60mm plates and<br />

exposed to a spectrum of AKAi-1 and Gemcitabine concentrations alone and<br />

in combination. Cell cycle was analyzed by propidium iodide staining and<br />

flow cytometry. Apoptosis was analyzed by PE-Annexin V staining and<br />

flow cytometry.<br />

Direct inhibition of constitutively active oncogenic Kras or its upstream<br />

regulators ineffective or impossible in clinical populations 3<br />

Targeted inhibition of downstream effectors of Kras may reveal new<br />

potential therapeutic targets<br />

RalA Activity: Active, phosphorylated RalA was quantified using a<br />

RalGTP assay (MilliporeTM ). Active RalA was pulled down by RalBP1 as<br />

per manufacturer’s instructions and quantified by western blot.<br />

Acknowledgements<br />

Hypotheses<br />

Targeting specific downstream effectors of Kras maybe an effective method<br />

of inhibiting Ras-driven PDA<br />

This program was supported by a research grant from the Howard Hughes Medical Institute and Whitman College.<br />

I would personally like to thank Dr. Sunil Hingorani, Dr. Vikas Goel, Amy Chang, Natalie Wong, Layken Fletcher, Dr.<br />

Paolo Provenzano, and Dr. Libing Feng for their help and guidance during this internship.<br />

AKAi-1<br />

- 0.5 1 2.5 5 - - - 0.5 0.5 1 1<br />

(µM)<br />

GEM<br />

- - - - - - 5 10 5 10 5 10<br />

(nM)<br />

Fig. 9: Effect of AKAi-1on apoptosis in KPC cells. Concentration-dependent increase in apoptosis was<br />

seen in AKAi-1-treated cells. Effect plateaued near IC50 value for AKAi-1 (2.06 µM). Concentrationdependent<br />

increase in apoptosis was also observed for Gemcitabine treatments. No additive or<br />

synergistic effects were observed from co-administration of AKAi-1 with Gemcitabine.<br />

Knockdown of KrasG12D by shRNA: Cells were infected with<br />

Doxycyclin-inducible TripZ vectors containing Kras shRNA. Knockdown<br />

efficiency was estimated by western blot.<br />

Using RalA to link Aurora Kinase A (AKA) and Kras validates AKA as a<br />

therapeutic target in treatment of PDA

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

Saved successfully!

Ooh no, something went wrong!