Summary

In Vivo Imaging to Measure Spontaneous Lung Metastasis of Orthotopically-injected Breast Tumor Cells

Published: June 23, 2022
doi:

Summary

The manuscript describes a methodology for the establishment as well as longitudinal growth monitoring of spontaneous lung metastasis from orthotopically-injected breast tumors, amenable to intervention at all stages of the metastatic cascade.

Abstract

Metastasis remains the primary cause of cancer-related death. The succession of events that characterize the metastatic cascade presents multiple opportunities for therapeutic intervention, and the ability to accurately model them in mice is critical to evaluate their effects. Here, a step-by-step protocol is presented for the establishment of orthotopic primary breast tumors and the subsequent monitoring of the establishment and growth of metastatic lesions in the lung using in vivo bioluminescence imaging. This methodology allows for the evaluation of treatment or its biological effects along the entire range of metastatic development, from primary tumor escape to outgrowth in the lungs. Breast orthotopic tumors are generated in mice via injection of a luciferase-labeled cell suspension in the 4th mammary gland. Tumors are allowed to grow and disseminate for a specific amount of time and are then surgically resected. Upon resection, spontaneous lung metastasis is detected, and the growth over time is monitored using in vivo bioluminescence imaging. At the desired experimental endpoint, lung tissue can be collected for downstream analysis. The treatment of established, clinically evident metastasis is critical to improve outcomes for stage IV cancer patients, and it can be evaluated through tail vein models of experimental lung metastasis. However, metastatic dissemination occurs early in breast cancer, and many patients have latent, subclinical disseminated disease after surgery. Utilization of spontaneous models such as this one provides the opportunity to study the whole spectrum of the disease, especially the systemic effects driven by treatment of the primary tumor such as pre-metastatic niche priming, and evaluate treatments on dormant and subclinical disease after surgery.

Introduction

Metastasis - the spread of cancer cells from the primary tumor to other parts of the body – remains the cause of death in more than 90% of cancer patients. This process is complex, involving migration of the tumor cells out of the primary tumor and intravasation into the circulation, survival in the blood, extravasation and survival in the target organ, re-instauration of the proliferative state, and outgrowth1. Spontaneous and transplantable murine cancer models have been used to investigate early or late stages of metastasis, each presenting its own advantages and disadvantages, which have been thoroughly discussed2,3,4.

Unlike previously thought, tumor cells abandon the primary tumor at early stages during tumor development, sometimes remaining dormant in distant tissues for what can be long periods of time5,6,7,8. In addition, there is mounting evidence of the strong systemic effects the primary tumor has on disease outcomes, often manifested through the secretion of soluble factors and exosomes that condition the metastatic soil or stimulate muscle wasting during cachexia9,10,11,12. For these reasons, modeling the length of the metastatic process in the initial presence of the primary tumor has become essential for achieving a more complete understanding of the biology driving these processes and testing potential new interventions aimed at disrupting or delaying the process.

In this work, a protocol is described for quantifying lung metastasis arising spontaneously from traceable cell lines injected orthotopically in the mammary gland, a process that models all of the above steps in the metastatic cascade. Transplantable models of metastasis are known to be more representative of human metastasis as compared to spontaneous, genetically-driven cancer models, thus improving clinical translatability2. Furthermore, this protocol utilizes bioluminescence imaging to study the growth and progression of spontaneous lung metastasis from primary breast tumors in real time within living animals, thus improving efficiency over more traditional histology-based assessments of metastatic dissemination. This protocol also includes evaluation of spontaneous metastasis after the surgical removal of the primary tumor, which is both clinically relevant and allows researchers to study the effect of minimal residual disease on the metastatic process. Finally, the use of immunocompetent mice confers the advantage of allowing for an intact immune system to shape the metastatic process, as is the case in human biology10,11.

Protocol

All animal procedures and protocols described here were approved by the Institutional Animal Care and Use Committee of Virginia Commonwealth University. 1. Preparation of cells for injection Thaw luciferase-transduced EO771 cells13 from liquid nitrogen storage and plate 1 x 106 cells in a 10 cm tissue-culture dish in complete cell culture medium (RPMI1640 + 10% FBS + 1% penicillin/streptomycin + 1% amphotericin B). Incub…

Representative Results

Orthotopic injection of mouse cancer cell lines into the 4th mammary fat pad of mice is a reproducible and reliable procedure for inducing mouse primary tumors. Utilizing the EO771 cell line transduced with luciferase in the conditions described in this protocol, primary tumors become palpable and can be measured using calipers about 7 days after injection and reach approximately 150 mm3 in volume at around 14 days following initial injection (Figure 1). Bioluminescence growth of …

Discussion

As the early nature of metastatic dissemination and the systemic effects of cancer become more widely recognized, the need for models in which both of these critical factors are taken into consideration becomes a necessity. This protocol allows researchers to monitor minimal residual disease and outgrowth of lung metastasis occurring spontaneously from primary breast tumors, accounting for the systemic effects of cancer that influence the metastatic process. Primary tumor removal is necessary to evaluate lung metastasis …

Disclosures

The authors have nothing to disclose.

Acknowledgements

Work in the Bos lab is supported by the Susan G. Komen Foundation (CCR18548205 P.D.B.), V Foundation (V2018-22 P.D.B.), and American Cancer Society (RSG-21-100-01-IBCD P.D.B.)

Materials

0.25% Trypsin/EDTA Hyclone SH30042.01
1.5ml microcentrifuge tubes USA Scientific 1615-5500
10% povidone-iodine solution Medline MDS093906
15ml centrifuge tubes VWR 89039-666
1X PBS Hyclone SH30256.01
28G 0.5ml U-100 Insulin Syringe BD Biosciences 329461
Amphotericin B Gemini Bio-products 400-104
Cautery Kit Braintree Scientific DEL2
D-Luciferin Potassium SydLabs MB102
Ethanol Koptec V1001
Fetal Bovine Serum R&D Systems S11150H
Forceps Fisherbrand 16-100-110
Growth factor-reduced Matrigel Corning 354230
Isoflurane Covetus 29405
IVIS Spectrum 200 Perkin Elmer 124262
Meloxicam (2mg/ml) Zoopharm LLC N/A By veterinary prescription
Penicillin/Streptomycin Gemini Bio-products 400-109
RPMI1640 Hyclone SH30027.01
Scissors Miltex 5-300
Silk sutures Braintree Scientific SUT-S 103
Surgical staples Reflex7 203-1000
Trypan Blue Gibco 15250-061

References

  1. Gupta, G. P., Massague, J. Cancer metastasis: Building a framework. Cell. 127 (4), 679-695 (2006).
  2. Bos, P. D., Nguyen, D. X., Massagué, J. Modeling metastasis in the mouse. Current Opinion in Pharmacology. 10 (5), 571-577 (2010).
  3. Francia, G., Cruz-Munoz, W., Man, S., Xu, P., Kerbel, R. S. Mouse models of advanced spontaneous metastasis for experimental therapeutics. Nature Reviews Cancer. 11 (2), 135-141 (2011).
  4. Gómez-Cuadrado, L., Tracey, N., Ma, R., Qian, B., Brunton, V. G. Mouse models of metastasis: Progress and prospects. Disease Models & Mechanisms. 10 (9), 1061-1074 (2017).
  5. Husemann, Y., Klein, C. A. The analysis of metastasis in transgenic mouse models. Transgenic Research. 18 (1), 1-5 (2009).
  6. Klein, C. A. Parallel progression of primary tumours and metastases. Nature Reviews Cancer. 9 (4), 302-312 (2009).
  7. Pantel, K., Brakenhoff, R. H. Dissecting the metastatic cascade. Nature Reviews Cancer. 4 (6), 448-456 (2004).
  8. Sosa, M. S., Bragado, P., Aguirre-Ghiso, J. A. Mechanisms of disseminated cancer cell dormancy: An awakening field. Nature Reviews Cancer. 14 (9), 611-622 (2014).
  9. Biswas, A. K., Acharyya, S. Understanding cachexia in the context of metastatic progression. Nature Reviews Cancer. 20 (5), 274-284 (2020).
  10. Liu, Y., Cao, X. Characteristics and significance of the pre-metastatic niche. Cancer Cell. 30 (5), 668-681 (2016).
  11. Peinado, H., et al. Pre-metastatic niches: Organ-specific homes for metastases. Nature Reviews Cancer. 17 (5), 302-317 (2017).
  12. Psaila, B., Lyden, D. The metastatic niche: Adapting the foreign soil. Nature Reviews Cancer. 9 (4), 285-293 (2009).
  13. Clark, N. M., et al. Regulatory T cells support breast cancer progression by opposing IFN-γ-dependent functional reprogramming of myeloid cells. Cell Reports. 33 (10), 108482 (2020).
  14. Liu, J., et al. Improved efficacy of neoadjuvant compared to adjuvant immunotherapy to eradicate metastatic disease. Cancer Discovery. 6 (12), 1382-1399 (2016).
  15. Thompson, A. M., Moulder-Thompson, S. L. Neoadjuvant treatment of breast cancer. Annals of Oncology. Official Journal of the European Society for Medical Oncology. 23, 231-236 (2012).
  16. Serganova, I., Blasberg, R. G. Molecular imaging with reporter genes: Has its promise been delivered. Journal of Nuclear Medicine: Official Publication, Society of Nuclear Medicine. 60 (12), 1665-1681 (2019).
  17. Grzelak, C. A., et al. Elimination of fluorescent protein immunogenicity permits modeling of metastasis in immune-competent settings. Cancer Cell. 40 (1), 1-2 (2022).
  18. Valiente, M., et al. Brain metastasis cell lines panel: A public resource of organotropic cell lines. 癌症研究. 80 (20), 4314-4323 (2020).
This article has been published
Video Coming Soon
Keep me updated:

.

Cite This Article
Sanon, S., Bos, P. D. In Vivo Imaging to Measure Spontaneous Lung Metastasis of Orthotopically-injected Breast Tumor Cells. J. Vis. Exp. (184), e64002, doi:10.3791/64002 (2022).

View Video