Summary

Detection of Lung Tumor Progression in Mice by Ultrasound Imaging

Published: February 27, 2020
doi:

Summary

This protocol describes the steps taken to induce KRAS lung tumors in mice as well as the quantification of formed tumors by ultrasound imaging. Small tumors are visualized in early timepoints as B-lines. At later timepoints, relative tumor volume measurements are achieved by the measurement tool in the ultrasound software.

Abstract

With ~1.6 million victims per year, lung cancer contributes tremendously to the worldwide burden of cancer. Lung cancer is partly driven by genetic alterations in oncogenes such as the KRAS oncogene, which constitutes ~25% of lung cancer cases. The difficulty in therapeutically targeting KRAS-driven lung cancer partly stems from having poor models that can mimic the progression of the disease in the lab. We describe a method that permits the relative quantification of primary KRAS lung tumors in a Cre-inducible LSL-KRAS G12D mouse model via ultrasound imaging. This method relies on brightness (B)-mode acquisition of the lung parenchyma. Tumors that are initially formed in this model are visualized as B-lines and can be quantified by counting the number of B-lines present in the acquired images. These would represent the relative tumor number formed on the surface of the mouse lung. As the formed tumors develop with time, they are perceived as deep clefts within the lung parenchyma. Since the circumference of the formed tumor is well-defined, calculating the relative tumor volume is achieved by measuring the length and width of the tumor and applying them in the formula used for tumor caliper measurements. Ultrasound imaging is a non-invasive, fast and user-friendly technique that is often used for tumor quantifications in mice. Although artifacts may appear when obtaining ultrasound images, it has been shown that this imaging technique is more advantageous for tumor quantifications in mice compared to other imaging techniques such as computed tomography (CT) imaging and bioluminescence imaging (BLI). Researchers can investigate novel therapeutic targets using this technique by comparing lung tumor initiation and progression between different groups of mice.

Introduction

As the leading cause of cancer-related deaths worldwide, lung cancer remains refractory to treatments, mainly due to lack of relevant pre-clinical models that can recapitulate the disease in the lab1. Around 25% of lung cancer cases are due to mutations in the KRAS oncogene2. KRAS-driven lung cancer is often associated with poor prognosis and low response to therapy, highlighting the importance of further studies in this disease2.

We optimized a method that allows the relative evaluation of lung tumor growth in real time in KRAS lung cancer-induced immune-competent mice. We use Lox-Stop-Lox KRAS G12D (LSL-KRAS G12D) mice in which the KRAS G12D oncogene can be expressed by Cre lentiviral vectors3,4. These vectors are driven by carbonic anhydrase 2, allowing the viral infection to take place specifically in alveolar epithelial cells5. In addition, to accelerate the initiation and progression of lung tumors, the lentiviral construct also expresses P53 shRNA from an U6/H1 promoter (the lentiviral construct herein will be referred to as Ca2Cre-shp53)6. The biological relevance of this method lies in the natural course of lung tumor development in mice as opposed to xenografts of non-orthotopic tumors in mice. An obstacle using the orthotopic method is monitoring lung tumor growth without sacrificing the mouse. To overcome this limitation, we optimized ultrasound imaging to permit the analysis of lung tumor progression in two-dimensional (2D) mode in this mouse model. Initiating tumors at 7 weeks post-infection are reflected as B-lines in ultrasound images, which can be counted, but will not reflect the exact number of tumors present on the lung. B-lines are characterized by laser-like vertical white lines arising from the pleural line in the lung parenchyma7,8. Large tumors can be visualized after 18 weeks of infection. The relative volume of these tumors is quantified by 2D measurements done on ultrasound.

This method is optimal for researchers investigating the effect of pharmacological drugs on lung tumor growth in the LSL-KRAS G12D mouse model. In addition, lung tumor progression can be compared between mice with different genetic lineages, to examine the importance of the presence or absence of certain genes/proteins on the development of lung tumor volume.

Protocol

Animal studies were performed in accordance with the Institutional Animal Care and Use Committee (IACUC) of McGill University and procedures were approved by the Animal Welfare Committee of McGill University (animal use protocol # 2009-5754). 1. Generation of CA2Cre-shp53 Lentiviral Titre NOTE: The following protocol is the same as that described in Xia et al.6, with minor modifications. Preparation of lentivirus (for 15 cm x 10 c…

Representative Results

After obtaining a lentiviral infectious titer of ~2 x 106 TU/mL (Figure 1), the Ca2Cre-shp53 lentivirus was intratracheally injected when LSL-KRAS G12D mice reached an appropriate age (6−8 weeks)9. Ultrasound imaging was performed after 7 weeks of infection upon initiation of tumors (Figure 3B). Imaging was done at 7 weeks in order to include the various types of precursor lesions that occur in the LSL-KR…

Discussion

We demonstrate a method that can assess lung tumor growth in the Cre-inducible LSL-KRAS G12D mouse model by ultrasound. This method can be used for evaluating the effect of pharmacological inhibitors on lung tumor growth. It can also be used to compare lung tumor growth between mice of different genetic backgrounds. Using this technique does not require specialized computational skills, however, it is important to be systematic in the number of frames used for analysis to allow for proper comparison if the method is used…

Disclosures

The authors have nothing to disclose.

Acknowledgements

We thank Dr. I. Verma for the lentiviral Ca2Cre-shp53 vector. The work was supported by funds from the Canadian Institutes of Health Research (CIHR MOP 137113) to AEK.

Materials

0.45 μm Acrodisc Syringe Filters Pall Corporation PN 4614
100-mm Cell Cultre Plate CELLSTAR 664 160
6-well Cell Culture Plate CELLSTAR 657 160
Amicon Ultra – 15 Centrifugal Filter Units Merck Millipore Ltd. UFC910024
BD LSR-Fortessa BD Biosciences 649225B 3024
CA2Cre-shp53 lentiviral vector From Dr. I Verma Laboratory
DMEM Multicell 319-005-CL
FBS Multicell 80450
LSL-KRASG12D mouse JAX Mice 8179
MX550S; Centre Transmit: 40 MHz FUJIFILM VisualSonics 51070
OptiMEM gibco 11058-021
Pen/strep Multicell 450-201-EL
pMD2.G Addgene 12259
PsPAX2 Addgene 12260
VEVO-3100 FUJIFILM VisualSonics 51072-50

References

  1. Eisenstein, M. Personalized medicine: Special treatment. Nature. 513, 8 (2014).
  2. Karachaliou, N., et al. KRAS mutations in lung cancer. Clinical Lung Cancer. 14 (3), 205-214 (2013).
  3. Jackson, E. L., et al. Analysis of lung tumor initiation and progression using conditional expression of oncogenic K-ras. Genes & Development. 15 (24), 3243-3248 (2001).
  4. DuPage, M., Dooley, A. L., Jacks, T. Conditional mouse lung cancer models using adenoviral or lentiviral delivery of Cre recombinase. Nature Protocol. 4 (7), 1064-1072 (2009).
  5. Chen, J., Lecuona, E., Briva, A., Welch, L. C., Sznajder, J. I. Carbonic anhydrase II and alveolar fluid reabsorption during hypercapnia. American Journal of Respiratory Cell and Molecular Biology. 38 (1), 32-37 (2008).
  6. Xia, Y., et al. Reduced cell proliferation by IKK2 depletion in a mouse lung-cancer model. Nature Cell Biology. 17 (4), 532 (2015).
  7. Demi, L., et al. Determination of a potential quantitative measure of the state of the lung using lung ultrasound spectroscopy. Scientific Reports. 7, 12746 (2017).
  8. Mohanty, K., et al. Characterization of the Lung Parenchyma Using Ultrasound Multiple Scattering. Ultrasound in Medicine and Biology. 43, 993-1003 (2017).
  9. Vandivort, T. C., An, D., Parks, W. C. An Improved Method for Rapid Intubation of the Trachea in Mice. Journal of Visualized Experiments. (108), e53771 (2016).
  10. Saraogi, A. Lung ultrasound: Present and future. Lung India. 32 (3), 250-257 (2015).
  11. Gargani, L., Volpicelli, G. How I do it: lung ultrasound. Cardiovascular Ultrasound. 12, 25 (2014).
  12. Soldati, G., et al. On the Physical Basis of Pulmonary Sonographic Interstitial Syndrome. Journal of Ultrasound in Medicine. 35 (10), 2975 (2016).
  13. Raes, F., et al. High-Resolution Ultrasound and Photoacoustic Imaging of Orthotopic Lung Cancer in Mice: New Perspectives for Onco-Pharmacology. PLoS One. 11 (4), 15 (2016).
  14. Lakshman, M., Needles, A. Screening and quantification of the tumor microenvironment with micro-ultrasound and photoacoustic imaging. Nature Methods. 12 (4), 372 (2015).
  15. Chichra, A., Makaryus, M., Chaudhri, P., Narasimhan, M. Ultrasound for the Pulmonary Consultant. Clinical Medicine Insights: Circulatory Respiratory and Pulmonary Medicine. 10, 9 (2016).
check_url/kr/60565?article_type=t

Play Video

Cite This Article
Ghaddar, N., Wang, S., Michaud, V., Kazimierczak, U., Ah-son, N., Koromilas, A. E. Detection of Lung Tumor Progression in Mice by Ultrasound Imaging. J. Vis. Exp. (156), e60565, doi:10.3791/60565 (2020).

View Video