Jan 1, 2024 · The first mouse metastasis model was the B16 melanoma model, which was developed from a spontaneous mouse tumor that metastasized to the lung and brain when reintroduced into syngeneic mice. 31 The B16 model laid the foundation for the seminal work of Fidler et al., which established many fundamental concepts of metastasis progression ... ... Furthermore, the use of experimental metastasis assays has also been highly successful for investigating the tissue specificity of metastases (for example, for experimental breast cancer metastases 65,66,70, using models that are also capable of spontaneous metastasis 71,72), as well as their response to targeted therapies 55 and the ... ... The second model is an experimental metastasis model. In this model, luciferase-labeled MDA-MB-231 human breast tumor cells are injected into the tail vein of NOD-SCID immunodeficient mice and the presence of tumor cells in mice will be detected through the noninvasive bioluminescence imaging ( 13 , 14 ). ... Clinically, metastasis may take decades to become manifest in certain types of cancer , and there is a dearth of mouse models to study the biology of the latent metastatic state . Both of these questions would benefit from the development of additional immunocompromised models (rev. in [ 57 ]). ... Jan 24, 2011 · Barnett, S. C. & Eccles, S. A. Studies of mammary carcinoma metastasis in a mouse model system. II: lectin binding properties of cells in relation to the incidence and organ distribution of ... ... Sep 1, 2017 · More recently, elegant mouse experiments again using experimental metastasis models have shown that tumor-derived exosomes (see Glossary, Box 1) induce pro-metastatic progenitor cells in the bone marrow through receptor tyrosine kinase MET signaling (Peinado et al., 2012), and that exosomal integrins (see Glossary, Box 1) can direct organ ... ... Oct 1, 2010 · Clinically, metastasis may take decades to become manifest in certain types of cancer [4], and there is a dearth of mouse models to study the biology of the latent metastatic state [2 ••]. Both of these questions would benefit from the development of additional immunocompromised models (reviewed in [ 57 ]). ... Mar 15, 2024 · Metastasis is a multistep process beginning with intravasation. In this step, cancer cells disseminate from their primary site into the blood stream. 2 In a consecutive step, cancer cells extravasate from the bloodstream into other organs, adapt to their new microenvironment, and grow to form metastasis. 3 Depending on the location of the primary tumor, different metastatic sites can be ... ... Oct 30, 2023 · The first mouse metastasis model was the B16 melanoma model, which was developed from a spontaneous mouse tumor that metastasized to the lung and brain when reintroduced into syngeneic mice.31 The B16 model laid the foundation for the seminal work of Fidler et al., which established many fundamental concepts of metastasis progression, including ... ... Dec 12, 2023 · Therefore, new mouse models of spinal metastasis need to be developed to simulate the process of spinal metastasis generation. Funding This work is supported by a grant from the Natural Science Foundation of Shanxi Province (No. 202204041101023). ... ">

An official website of the United States government

Official websites use .gov A .gov website belongs to an official government organization in the United States.

Secure .gov websites use HTTPS A lock ( Lock Locked padlock icon ) or https:// means you've safely connected to the .gov website. Share sensitive information only on official, secure websites.

  • Publications
  • Account settings
  • Advanced Search
  • Journal List

NIHPA Author Manuscripts logo

Modeling metastasis in the mouse

Paula d bos, don x nguyen, joan massagué.

  • Author information
  • Article notes
  • Copyright and License information

Correspondence: Joan Massagué, Box 116, Memorial Sloan-Kettering Cancer Center, 1275 York Avenue, New York, NY 10065 USA, Phone: 646-888-2044, [email protected]

Present address: Immunology Program, Memorial Sloan-Kettering Cancer Center, New York, NY, USA

Issue date 2010 Oct.

Metastasis is a complex clinical and biological problem presently under intense study, and several model systems are in use to experimentally recapitulate and dissect the various steps of the metastatic process. Genetically engineered mouse models provide faithful renditions of events in tumor progression, angiogenesis, and local invasion that set the stage for metastasis, whereas engrafting of human or mouse tumor tissues into mouse hosts has been successfully exploited to investigate metastatic dissemination and colonization of distant organs. Real-time, high-resolution microscopy in live animals, and comprehensive genetic and molecular profiling are effective tools to interrogate diverse metastatic cancer cell phenotypes as well as the metastatic tumor microenvironment in different organs. By integrating the information obtained with these complementary approaches the field is currently obtaining an unprecedented level of understanding of the biology, molecular basis, and therapeutic vulnerabilities of metastasis.

Introduction

Metastasis is a challenging clinical problem and the cause of most deaths from cancer. As a biological process, metastasis is quite complex as it reflects the many barriers that cancer cells that leave a primary tumor must overcome to generate aggressive secondary lesions [ 1 ]. Depending on the cancer type, metastasis may be achieved by just a rare minority of tumor-initiating cells that reach, survive and eventually overtake a distant tissue microenvironment over a long period of time, as in certain types of breast cancer [ 2 , 3 ], or it may represent a relatively common occurrence among primary tumor cell populations that are primed to perform many of the necessary steps and prone to forming rapidly growing metastatic colonies as, for example, in lung adenocarcinoma [ 4 , 5 ].

Certain fundamental properties of metastatic cells, including migration and invasiveness, have been the subject of many studies using a variety of in vitro model systems. Technological advances such as fluorescent or bioluminescent reporter molecules and sophisticated microscopy have allowed sensitive and accurate analysis of these processes and their molecular underpinnings at the single-cell or cell-cluster levels [ 6 , 7 ]. These experimental systems additionally provide comparatively inexpensive methods of choice to screen RNAi, cDNA or chemical libraries for mediators or inhibitors of these cancer cell functions [ 8 – 10 ]. In vitro model systems have contributed to define the role of candidate metastasis genes in particular steps of the metastatic cascade [ 11 , 12 ]. However, these models can provide surrogate systems for the analysis of only a limited set of events in the metastatic cascade, which in vivo involves multiple steps within specific tissue contexts.

To better understand tumor development and progression in vivo , two general strategies have been pursued in mice: genetically engineered models of cancer, and transplantable tumor model systems. These strategies provide complementary approaches to the dissection of specific metastatic steps. Cancer models driven by the introduction of oncogenic mutations in a tissue-specific manner can faithfully recapitulate important aspects of tumor initiation, local progression, and response to therapy [ 13 , 14 ]. In these models, cancer develops with high penetrance in a stepwise manner, enabling the study of tumor initiation and early steps of metastatic dissemination ( Figure 1 ). However, metastasis in these models is often restricted to lymph nodes and the lungs, or is missing altogether ( Table 1 ). Syngeneic and xenograft models in which mouse or human cancer cells are introduced into immunocompatible or immunocompromised mice provide at present methods of choice to experimentally address metastatic dissemination to, and colonization of relevant organs. Syngeneic models enable the study of the complete microenvironmental interface in the mouse but are limited to study mouse cancer cell metastasis. Conversely, in spite of the caveat of an incomplete immune system, xenograft models provide a superior alternative to the study of metastasis of human cancer cells in vivo .

Figure 1

Contribution of different mouse models to the various steps of metastatic dissemination. GEM: Genetically engineered mouse models, GRAFTS: xeno- or allograft transplantation.

Metastatic patterns of dissemination

Here we present an overview of the advantages and disadvantages of the principal mouse model systems currently used for the study of metastasis ( Table 2 ), and how the combined use of these systems complemented with in vitro models is yielding an increasingly robust understanding of the multiple modes and steps of metastasis.

Advantages and disadvantages of genetically engineered and transplantable models for the study of metastasis

Modeling early metastatic steps

Cancer cells in a primary tumor have already acquired a number of aggressive functions that will remain important throughout the rest of their metastatic progression. These functions generally include motility, invasiveness, resistance to hypoxia and reactive oxygen species, survival after detachment, and evasion of immune surveillance. The accumulation of these properties in human cancer cells is thought to occur in the context of the primary tumor, and genetically engineered tumors in mice provide powerful models to delineate the acquisition of these functions ( Figure 1 ).

It has been proposed that secondary tumor formation involves rare cell variants that have accumulated a complete set of genetic mutations in the primary tumor that enables these cells to grow in a distant organ [ 15 ]. However, this hypothesis has been challenged based on the detection of widespread gene expression patterns in primary tumors that strongly predict metastatic competence [ 16 ], and the analysis of growth dynamics of human breast primary tumors and metastases [ 17 ]. Genetically marked transplantable tumors in mouse mammary carcinoma models were used to demonstrate that cancer cells can disseminate at the premalignant stage [ 18 , 19 ]. Moreover, ex vivo genetic manipulation of tumor-derived cells prior to implantation in cleared mouse mammary fat pads was used to identify genes that influence this process [ 19 ]. Other transplantation methods using inducible genetically engineered oncogenes have shown that non-transformed mammary cells introduced in the mouse circulation can extravasate in the lungs, and give rise to tumor foci in the lung parenchyma upon oncogene induction [ 20 ]. However, the question of whether early disseminated tumor cells (DTCs) are responsible for the outgrowth of secondary tumors remains a matter of debate. In one approach, early-stage mouse tumors were transplanted for variable time periods, and metastatic outgrowth measured upon resection after a fixed number of weeks. The results suggested that early DTCs are inefficient at initiating secondary tumors [ 19 ].

Xenografting of limiting dilutions of cancer cells are standard for evaluating tumor-initiating capacity [ 21 ], and has been instrumental in linking epithelial-mesenchymal trans-differentiation (EMT) to tumor-initiating “cancer stem” cell phenotype in breast cancer [ 22 ]. A recently introduced interleukin-2 receptor-deficient NOD/SCID mouse strain has been used to demonstrate that one single human melanoma cell can develop into a lethal, metastatic tumor [ 5 ]. Tumor engrafting models are extensively used to delineate the role of genes of interest in early metastatic steps at orthotopic sites. Both, xeno- and allo-transplantation have been successfully used to delineate the role of genes and miRNAs in the invasive capacity of tumor cells [ 12 , 23 – 25 ], and to dissect the distinct contributions of particular genes in the context of mammary tumors versus the context of lung metastasis [ 11 , 26 ].

The importance of the microenvironment in tumor progression is widely recognized, and genetically engineered mouse models have provided key insights into the relevance of the tumor microenvironment in metastasis [ 27 ]. Dissenting the contribution of different components of the tumor stroma represent a significant experimental challenge. Nonetheless, progress has been made with the use of bone marrow transplantation from genetically engineered donor mice. Such bone marrow transplants into tumor bearing animals can shed light into the role of a candidate metastasis gene expressed in either the cancer cells or a particular cell type in the tumor microenvironment. Recent examples of this approach have revealed the specific contribution of macrophage-derived cathepsins in pancreatic cancer and breast cancer cell invasion [ 28 ]. Similar genetic manipulations in a double transgenic PyMT/RAG1−/− breast cancer model have implicated CD4 T effector cells in tumor cell invasion and metastasis [ 29 ]. This methodology is highly promising to define the role of the innate and adaptive immune system in tumor cell dissemination.

Metastatic dissemination and colonization: transplantable tumor models

Establishment of secondary tumors imposes different demands on disseminated cancer cells depending on the target organ. Xenograft models provide an effective system to investigate secondary organ colonization of human cells, and remain the model of choice for pre-clinical studies of human tumor-derived cells ( Table 3 ). Intracardiac inoculation of cancer cells into the arterial circulation of mice allows the systemic distribution of these cells to all organs for the analysis of metastatic functions including organ-specific extravasation, survival in the newly invaded parenchyma, retention of tumor-reinitiating capacity, and overt colonization [ 30 ]. In contrast, tail-vein inoculation forces cancer cells to lodge in lung capillaries, which allows an assessment of lung extravasation and colonization functions [ 31 ]. Carotid artery inoculation likewise targets cancer cells to the brain [ 32 ].

Sources and utilization of clinical material.

In vivo selection of organ-specific metastatic variants from human malignant samples and cell lines, coupled with analysis of mRNA and microRNA expression patters has allowed the identification of organ-specific metastasis genes and functions. By comparing the results of this type of analysis with clinical gene expression data sets, it is possible to identify metastasis-associated genes of clinical relevance. Several gene sets have been identified in this manner that are associated with organ-specific relapse in breast cancer patients [ 33 , 34 ]. This information in turn can be used to guide functional studies for the discovery of genes that mediate metastasis, including genes that prime cancer cells for extravasation across the tight endothelial walls in the lungs or the brain [ 11 , 34 ]. Variants of this approach have identified new mediators of circulating cancer cell interaction with vascular capillary walls [ 35 ], and genes that support the tumor-reinitiating capacity of disseminated cancer cells in the lung parenchyma [ 36 ].

Another approach is based on interrogating clinical gene expression data sets for associations between specific pathways and particular disease outcomes. By combining this information with functional assays it was recently shown that a hyperactive Wnt pathway in lung adenocarcinoma tumors supports aggressive multi-organ metastasis to brain and bones [ 37 ], whereas a high level of Src activity in breast tumors endows disseminating cancer cells with an enhanced capacity to survival in the bone marrow microenvironment and may contribute to late-onset bone metastasis [ 2 ].

Transplantation studies have also illuminated other aspects of metastasis including the systemic effect of transplanted tumors. Allografted tumors can cause the mobilization of VEGFR1 + bone marrow-derived cells to the lungs for the establishment of a “pre-metastatic niche” to host incoming cancer cells [ 38 ]. The use of xenografts has also allowed a demonstration that an indolent tumor can be stimulated to grow by bone marrow-derived cells mobilized by systemic signals from a separate tumor [ 39 ]. Inducible RNAi technology in a syngeneic model of lung metastasis has shown that recruitment of endothelial progenitor cells is essential for the angiogenic switch that facilitates macrometastatic growth [ 40 ].

A self-seeding mechanism by which circulating cancer cells reinfiltrate and populate the tumor of origin has been proposed to explain certain aspects of tumor growth and metastatic population dynamics [ 41 ]. A recent experimental demonstration of this phenomenon employed xenograft and allograft models of orthotopic tumor seeding by cancer cells entering the circulation from a separate tumor mass, from lung metastatic nodules, or from direct intra-arterial inoculation [ 42 ].

Although genetically engineered mouse models provide good systems for the pre-clinical evaluation of therapeutic agents [ 43 , 44 ], the response of human cancer cells to therapy in vivo requires the use of xenograft models. Of particular relevance is the xenografting of metastatic cell lines in orthotopic locations, followed by resection of the primary tumors and initiation of therapy. This set up approximates the situation observed in patients with advanced disease [ 45 ].

Visualizing metastasis

Tracking cancer cells in real time in whole animals has provided a tremendous advantage in the dynamic monitoring of metastatic development. Of particular relevance, multimodality imaging technology such as the triple fusion protein reporter with herpes simplex virus 1 thymidine kinase (HSV1-TK), fused to enhanced green fluorescent protein (eGFP), and firefly luciferase has enabled the use of nuclear imaging, bioluminiscence and fluorescence imaging in a single experiment. In addition, the same tissues can be further analyzed by histological detection of the fluorescence GFP in frozen sections, or immunohistochemical detection of the reporter [ 46 ]. Variants of these reporters with different emission wavelengths, duration of the signal, and split versions for complementation studies exist that are useful in different applications [ 47 ]. The development of more advanced reporter systems based on similar technology has also permitted the in vivo monitoring of gene pathway activity and inhibition [ 48 ].

Intravital microscopy is a key resource to visualize cancer cells performing various metastatic steps in situ (reviewed in [ 49 ]). Macrophage-assisted tumor cell migration involving an autocrine loop has relied on the use of this approach for the analysis of polyoma middle-T transgenic mammary tumors in mice [ 50 ]. In this assay, a fine needle containing a chemoattractant is introduced into the tumor, and migrating cells can be recovered for further analysis. The use of this technology has also shed light on the interaction between migrating cancer cells and macrophages (reviewed in [ 51 ]).

Long-term spinning disk confocal microscopy represents another application of advanced microscopy and improved tracing techniques to the examination of cellular movements and interactions in the tumor microenvironment. This imaging technique enables the dynamic visualization of stromal cells in defined tumor areas, and its combination with fluorescent reporter knock-in mice in which different immune cell types are marked, or injection of fluorescent antibodies or dextrans, enables the rapid collection of images for studying behavior of moving cells [ 52 ]. Rapid and prolonged visualization of tumor growth parameters such as angiogenesis, lymphangiogenesis, tissue viability, and response to therapy in larger tumor areas have also been achieved by the introduction of optical frequency domain imaging. This approach has the advantage of not requiring labeling or contrasting agents [ 53 ].

Multiphoton laser scanning microscopy has enabled live imaging of challenging metastatic sites like the brain. The use of cranial windows and fluorescently labeled tumor cells and dextrans has allowed a visualization of brain metastatic cells closely interacting with the microvasculature [ 54 ]. A longer term imaging study using genetic labeling of the vasculature has made possible to image several steps in the process of brain metastasis, demonstrating active extravasation of tumors cells into the brain, perivascular growth, and responses to therapy [ 55 ]. Advances in the visualization of protease activity such as the use of ACPP (activatable cell penetrating peptide) has also enabled the in vivo labeling of MMP protease activity in xenograft and genetically engineered tumors and small metastatic foci in the lungs [ 56 ]. The rapidly evolving field of intravital microscopy is likely to provide in the future new means to analyze metastatic behavior and performing detailed functional analysis of genetically engineered cells.

Conclusions and perspectives

A great deal of progress has been made in the study of the various metastatic steps in the past few years. However, many clinically relevant questions remain unanswered, owing partly to a lack of suitable animal models. The problem of metastatic dormancy and the role of the immune system in metastasis are prominent in this regard. Clinically, metastasis may take decades to become manifest in certain types of cancer [ 4 ], and there is a dearth of mouse models to study the biology of the latent metastatic state [ 2 ]. Both of these questions would benefit from the development of additional immunocompromised models (rev. in [ 57 ]). Progress in this direction is being made with human tissues implanted in the mouse to serve as recipients for human cancer cells [ 58 , 59 ]. Such systems may provide important new models for pre-clinical studies of anti-metastatic agents. Similarly, the ability to uncouple primary tumor growth from specific metastatic steps is a coveted feature of genetically engineered mouse models of cancer. The use of reversibly inducible oncogenic alterations [ 60 – 62 ], and stable [ 63 ] or reversible [ 64 ] RNAi targeting of genes of interest represents only some of the new tools that can be applied to the analysis of metastatic progression in genetically engineered mouse models. The development of new and improved experimental metastasis methods, and a better integration of the results with clinically data promises to support a sustained expansion of our ability to understand and fight metastasis.

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

  • 1. Gupta GP, Massague J. Cancer metastasis: building a framework. Cell. 2006;127(4):679–695. doi: 10.1016/j.cell.2006.11.001. [ DOI ] [ PubMed ] [ Google Scholar ]
  • 2. Zhang XH, Wang Q, Gerald W, Hudis CA, Norton L, Smid M, Foekens JA, Massague J. Latent bone metastasis in breast cancer tied to Src-dependent survival signals. Cancer Cell. 2009;16(1):67–78. doi: 10.1016/j.ccr.2009.05.017.. ••This article (and ref. 37 ) pioneered the analysis of clinical gene expression analysis with xenograft models of metastasis to identify signaling pathways involved in metastatic dissemination.
  • 3. Pantel K, Brakenhoff RH. Dissecting the metastatic cascade. Nat Rev Cancer. 2004;4(6):448–456. doi: 10.1038/nrc1370. [ DOI ] [ PubMed ] [ Google Scholar ]
  • 4. Nguyen DX, Bos PD, Massague J. Metastasis: from dissemination to organ-specific colonization. Nat Rev Cancer. 2009;9(4):274–284. doi: 10.1038/nrc2622. [ DOI ] [ PubMed ] [ Google Scholar ]
  • 5. Quintana E, Shackleton M, Sabel MS, Fullen DR, Johnson TM, Morrison SJ. Efficient tumour formation by single human melanoma cells. Nature. 2008;456(7222):593–598. doi: 10.1038/nature07567. [ DOI ] [ PMC free article ] [ PubMed ] [ Google Scholar ]
  • 6. Menon MB, Ronkina N, Schwermann J, Kotlyarov A, Gaestel M. Fluorescence-based quantitative scratch wound healing assay demonstrating the role of MAPKAPK-2/3 in fibroblast migration. Cell Motility and the Cytoskeleton. 2009;66(12):1041–1047. doi: 10.1002/cm.20418. 10.1002/cm.20418. [ DOI ] [ PubMed ] [ Google Scholar ]
  • 7. Ghajar CM, Bissell MJ. Extracellular matrix control of mammary gland morphogenesis and tumorigenesis: insights from imaging. Histochem Cell Biol. 2008;130(6):1105–1118. doi: 10.1007/s00418-008-0537-1. [ DOI ] [ PMC free article ] [ PubMed ] [ Google Scholar ]
  • 8. Gobeil S, Zhu X, Doillon CJ, Green MR. A genome-wide shRNA screen identifies GAS1 as a novel melanoma metastasis suppressor gene. Genes Dev. 2008;22(21):2932–2940. doi: 10.1101/gad.1714608. [ DOI ] [ PMC free article ] [ PubMed ] [ Google Scholar ]
  • 9. Huang Q, Gumireddy K, Schrier M, le Sage C, Nagel R, Nair S, Egan DA, Li A, Huang G, Klein-Szanto AJ, Gimotty PA, et al. The microRNAs miR-373 and miR-520c promote tumour invasion and metastasis. Nat Cell Biol. 2008;10(2):202–210. doi: 10.1038/ncb1681. [ DOI ] [ PubMed ] [ Google Scholar ]
  • 10. Douma S, Van Laar T, Zevenhoven J, Meuwissen R, Van Garderen E, Peeper DS. Suppression of anoikis and induction of metastasis by the neurotrophic receptor TrkB. Nature. 2004;430(7003):1034–1039. doi: 10.1038/nature02765. [ DOI ] [ PubMed ] [ Google Scholar ]
  • 11. Gupta GP, Nguyen DX, Chiang AC, Bos PD, Kim JY, Nadal C, Gomis RR, Manova-Todorova K, Massague J. Mediators of vascular remodelling co-opted for sequential steps in lung metastasis. Nature. 2007;446(7137):765–770. doi: 10.1038/nature05760. [ DOI ] [ PubMed ] [ Google Scholar ]
  • 12. Valastyan S, Reinhardt F, Benaich N, Calogrias D, Szasz AM, Wang ZC, Brock JE, Richardson AL, Weinberg RA. A pleiotropically acting microRNA, miR-31, inhibits breast cancer metastasis. Cell. 2009;137(6):1032–1046. doi: 10.1016/j.cell.2009.03.047. [ DOI ] [ PMC free article ] [ PubMed ] [ Google Scholar ] [ Retracted ]
  • 13. Jonkers J, Berns A. Conditional mouse models of sporadic cancer. Nat Rev Cancer. 2002;2(4):251–265. doi: 10.1038/nrc777. [ DOI ] [ PubMed ] [ Google Scholar ]
  • 14. Frese KK, Tuveson DA. Maximizing mouse cancer models. Nat Rev Cancer. 2007;7(9):645–658. doi: 10.1038/nrc2192. [ DOI ] [ PubMed ] [ Google Scholar ]
  • 15. Fidler IJ. The pathogenesis of cancer metastasis: the 'seed and soil' hypothesis revisited. Nat Rev Cancer. 2003;3(6):453–458. doi: 10.1038/nrc1098. [ DOI ] [ PubMed ] [ Google Scholar ]
  • 16. Bernards R, Weinberg RA. A progression puzzle. Nature. 2002;418(6900):823. doi: 10.1038/418823a. [ DOI ] [ PubMed ] [ Google Scholar ]
  • 17. Klein CA. Parallel progression of primary tumours and metastases. Nat Rev Cancer. 2009;9(4):302–312. doi: 10.1038/nrc2627. [ DOI ] [ PubMed ] [ Google Scholar ]
  • 18. Husemann Y, Geigl JB, Schubert F, Musiani P, Meyer M, Burghart E, Forni G, Eils R, Fehm T, Riethmuller G, Klein CA. Systemic spread is an early step in breast cancer. Cancer Cell. 2008;13(1):58–68. doi: 10.1016/j.ccr.2007.12.003. [ DOI ] [ PubMed ] [ Google Scholar ]
  • 19. Kouros-Mehr H, Bechis SK, Slorach EM, Littlepage LE, Egeblad M, Ewald AJ, Pai SY, Ho IC, Werb Z. GATA-3 links tumor differentiation and dissemination in a luminal breast cancer model. Cancer Cell. 2008;13(2):141–152. doi: 10.1016/j.ccr.2008.01.011.. ••Using a mouse mammary tumor model and sofisticated hyperplasia transplantation techniques, the authors demonstrate the timewise dissemination and metastatic development, and demosntrate the role of GATA3 in the process.
  • 20. Podsypanina K, Du YC, Jechlinger M, Beverly LJ, Hambardzumyan D, Varmus H. Seeding and propagation of untransformed mouse mammary cells in the lung. Science. 2008;321(5897):1841–1844. doi: 10.1126/science.1161621. [ DOI ] [ PMC free article ] [ PubMed ] [ Google Scholar ]
  • 21. Al-Hajj M, Wicha MS, Benito-Hernandez A, Morrison SJ, Clarke MF. Prospective identification of tumorigenic breast cancer cells. Proc Natl Acad Sci U S A. 2003;100(7):3983–3988. doi: 10.1073/pnas.0530291100. [ DOI ] [ PMC free article ] [ PubMed ] [ Google Scholar ]
  • 22. Mani SA, Guo W, Liao MJ, Eaton EN, Ayyanan A, Zhou AY, Brooks M, Reinhard F, Zhang CC, Shipitsin M, Campbell LL, et al. The epithelial-mesenchymal transition generates cells with properties of stem cells. Cell. 2008;133(4):704–715. doi: 10.1016/j.cell.2008.03.027. [ DOI ] [ PMC free article ] [ PubMed ] [ Google Scholar ]
  • 23. Tavazoie SF, Alarcon C, Oskarsson T, Padua D, Wang Q, Bos PD, Gerald WL, Massague J. Endogenous human microRNAs that suppress breast cancer metastasis. Nature. 2008;451(7175):147–152. doi: 10.1038/nature06487. [ DOI ] [ PMC free article ] [ PubMed ] [ Google Scholar ]
  • 24. Gumireddy K, Li A, Gimotty PA, Klein-Szanto AJ, Showe LC, Katsaros D, Coukos G, Zhang L, Huang Q. KLF17 is a negative regulator of epithelial-mesenchymal transition and metastasis in breast cancer. Nat Cell Biol. 2009;11(11):1297–1304. doi: 10.1038/ncb1974. [ DOI ] [ PMC free article ] [ PubMed ] [ Google Scholar ]
  • 25. Ma L, Young J, Prabhala H, Pan E, Mestdagh P, Muth D, Teruya-Feldstein J, Reinhardt F, Onder TT, Valastyan S, Westermann F, et al. miR-9, a MYC/MYCN-activated microRNA, regulates E-cadherin and cancer metastasis. Nat Cell Biol. 2010;12(3):247–256. doi: 10.1038/ncb2024. [ DOI ] [ PMC free article ] [ PubMed ] [ Google Scholar ]
  • 26. Padua D, Zhang XH, Wang Q, Nadal C, Gerald WL, Gomis RR, Massague J. TGFbeta primes breast tumors for lung metastasis seeding through angiopoietin-like 4. Cell. 2008;133(1):66–77. doi: 10.1016/j.cell.2008.01.046. [ DOI ] [ PMC free article ] [ PubMed ] [ Google Scholar ]
  • 27. Joyce JA, Pollard JW. Microenvironmental regulation of metastasis. Nat Rev Cancer. 2009;9(4):239–252. doi: 10.1038/nrc2618. [ DOI ] [ PMC free article ] [ PubMed ] [ Google Scholar ]
  • 28. Gocheva V, Wang HW, Gadea BB, Shree T, Hunter KE, Garfall AL, Berman T, Joyce JA. IL-4 induces cathepsin protease activity in tumor-associated macrophages to promote cancer growth and invasion. Genes Dev. 2010;24(3):241–255. doi: 10.1101/gad.1874010.. ••See ref. 29 .
  • 29. DeNardo DG, Barreto JB, Andreu P, Vasquez L, Tawfik D, Kolhatkar N, Coussens LM. CD4(+) T cells regulate pulmonary metastasis of mammary carcinomas by enhancing protumor properties of macrophages. Cancer Cell. 2009;16(2):91–102. doi: 10.1016/j.ccr.2009.06.018.. ••References 28 and 29 demonstrate the power of bone marrow transplantation technology to uncover the role of stromal components in cancer progression.
  • 30. Yin JJ, Selander K, Chirgwin JM, Dallas M, Grubbs BG, Wieser R, Massague J, Mundy GR, Guise TA. TGF-beta signaling blockade inhibits PTHrP secretion by breast cancer cells and bone metastases development. J Clin Invest. 1999;103(2):197–206. doi: 10.1172/JCI3523. [ DOI ] [ PMC free article ] [ PubMed ] [ Google Scholar ]
  • 31. Hart IR, Fidler IJ. Role of organ selectivity in the determination of metastatic patterns of B16 melanoma. Cancer Res. 1980;40(7):2281–2287. [ PubMed ] [ Google Scholar ]
  • 32. Lorger M, Felding-Habermann B. Capturing Changes in the Brain Microenvironment during Initial Steps of Breast Cancer Brain Metastasis. Am J Pathol. 2010 doi: 10.2353/ajpath.2010.090838. [ DOI ] [ PMC free article ] [ PubMed ] [ Google Scholar ]
  • 33. Minn AJ, Gupta GP, Siegel PM, Bos PD, Shu W, Giri DD, Viale A, Olshen AB, Gerald WL, Massague J. Genes that mediate breast cancer metastasis to lung. Nature. 2005;436(7050):518–524. doi: 10.1038/nature03799.. ••See ref. 34
  • 34. Bos PD, Zhang XH, Nadal C, Shu W, Gomis RR, Nguyen DX, Minn AJ, van de Vijver MJ, Gerald WL, Foekens JA, Massague J. Genes that mediate breast cancer metastasis to the brain. Nature. 2009;459(7249):1005–1009. doi: 10.1038/nature08021.. ••References 33 and 34 have pioneered the combined analysis of xenograft-derived and clinical sample gene expression data to identify organ-specific genes.
  • 35. Brown DM, Ruoslahti E. Metadherin, a cell surface protein in breast tumors that mediates lung metastasis. Cancer Cell. 2004;5(4):365–374. doi: 10.1016/s1535-6108(04)00079-0. [ DOI ] [ PubMed ] [ Google Scholar ]
  • 36. Gupta GP, Perk J, Acharyya S, de Candia P, Mittal V, Todorova-Manova K, Gerald WL, Brogi E, Benezra R, Massague J. ID genes mediate tumor reinitiation during breast cancer lung metastasis. Proc Natl Acad Sci U S A. 2007;104(49):19506–19511. doi: 10.1073/pnas.0709185104. [ DOI ] [ PMC free article ] [ PubMed ] [ Google Scholar ]
  • 37. Nguyen DX, Chiang AC, Zhang XH, Kim JY, Kris MG, Ladanyi M, Gerald WL, Massague J. WNT/TCF signaling through LEF1 and HOXB9 mediates lung adenocarcinoma metastasis. Cell. 2009;138(1):51–62. doi: 10.1016/j.cell.2009.04.030.. ••See ref. 2
  • 38. Kaplan RN, Riba RD, Zacharoulis S, Bramley AH, Vincent L, Costa C, MacDonald DD, Jin DK, Shido K, Kerns SA, Zhu Z, et al. VEGFR1-positive haematopoietic bone marrow progenitors initiate the pre-metastatic niche. Nature. 2005;438(7069):820–827. doi: 10.1038/nature04186. [ DOI ] [ PMC free article ] [ PubMed ] [ Google Scholar ]
  • 39. McAllister SS, Gifford AM, Greiner AL, Kelleher SP, Saelzler MP, Ince TA, Reinhardt F, Harris LN, Hylander BL, Repasky EA, Weinberg RA. Systemic endocrine instigation of indolent tumor growth requires osteopontin. Cell. 2008;133(6):994–1005. doi: 10.1016/j.cell.2008.04.045. [ DOI ] [ PMC free article ] [ PubMed ] [ Google Scholar ]
  • 40. Gao D, Nolan DJ, Mellick AS, Bambino K, McDonnell K, Mittal V. Endothelial progenitor cells control the angiogenic switch in mouse lung metastasis. Science. 2008;319(5860):195–198. doi: 10.1126/science.1150224. [ DOI ] [ PubMed ] [ Google Scholar ]
  • 41. Norton L, Massague J. Is cancer a disease of self-seeding? Nat Med. 2006;12(8):875–878. doi: 10.1038/nm0806-875. [ DOI ] [ PubMed ] [ Google Scholar ]
  • 42. Kim MY, Oskarsson T, Acharyya S, Nguyen DX, Zhang XH, Norton L, Massague J. Tumor self-seeding by circulating cancer cells. Cell. 2009;139(7):1315–1326. doi: 10.1016/j.cell.2009.11.025. [ DOI ] [ PMC free article ] [ PubMed ] [ Google Scholar ]
  • 43. De Palma M, Mazzieri R, Politi LS, Pucci F, Zonari E, Sitia G, Mazzoleni S, Moi D, Venneri MA, Indraccolo S, Falini A, et al. Tumor-targeted interferon-alpha delivery by Tie2-expressing monocytes inhibits tumor growth and metastasis. Cancer Cell. 2008;14(4):299–311. doi: 10.1016/j.ccr.2008.09.004. [ DOI ] [ PubMed ] [ Google Scholar ]
  • 44. Paez-Ribes M, Allen E, Hudock J, Takeda T, Okuyama H, Vinals F, Inoue M, Bergers G, Hanahan D, Casanovas O. Antiangiogenic therapy elicits malignant progression of tumors to increased local invasion and distant metastasis. Cancer Cell. 2009;15(3):220–231. doi: 10.1016/j.ccr.2009.01.027. [ DOI ] [ PMC free article ] [ PubMed ] [ Google Scholar ]
  • 45. Ebos JM, Lee CR, Cruz-Munoz W, Bjarnason GA, Christensen JG, Kerbel RS. Accelerated metastasis after short-term treatment with a potent inhibitor of tumor angiogenesis. Cancer Cell. 2009;15(3):232–239. doi: 10.1016/j.ccr.2009.01.021.. ••The authors demonstrate acceleration of metastasis upon anti-angiogenic therapies. Importantly, the authors mimmick clinical situations by removing orthotopically growing tumors before starting the therapy.
  • 46. Ponomarev V, Doubrovin M, Serganova I, Vider J, Shavrin A, Beresten T, Ivanova A, Ageyeva L, Tourkova V, Balatoni J, Bornmann W, et al. A novel triple-modality reporter gene for whole-body fluorescent, bioluminescent, and nuclear noninvasive imaging. Eur J Nucl Med Mol Imaging. 2004;31(5):740–751. doi: 10.1007/s00259-003-1441-5. [ DOI ] [ PubMed ] [ Google Scholar ]
  • 47. Dothager RS, Flentie K, Moss B, Pan MH, Kesarwala A, Piwnica-Worms D. Advances in bioluminescence imaging of live animal models. Curr Opin Biotechnol. 2009;20(1):45–53. doi: 10.1016/j.copbio.2009.01.007. [ DOI ] [ PMC free article ] [ PubMed ] [ Google Scholar ]
  • 48. Korpal M, Yan J, Lu X, Xu S, Lerit DA, Kang Y. Imaging transforming growth factor-beta signaling dynamics and therapeutic response in breast cancer bone metastasis. Nat Med. 2009;15(8):960–966. doi: 10.1038/nm.1943. [ DOI ] [ PubMed ] [ Google Scholar ]
  • 49. Condeelis J, Segall JE. Intravital imaging of cell movement in tumours. Nat Rev Cancer. 2003;3(12):921–930. doi: 10.1038/nrc1231. [ DOI ] [ PubMed ] [ Google Scholar ]
  • 50. Wyckoff JB, Wang Y, Lin EY, Li JF, Goswami S, Stanley ER, Segall JE, Pollard JW, Condeelis J. Direct visualization of macrophage-assisted tumor cell intravasation in mammary tumors. Cancer Res. 2007;67(6):2649–2656. doi: 10.1158/0008-5472.CAN-06-1823. [ DOI ] [ PubMed ] [ Google Scholar ]
  • 51. Qian BZ, Pollard JW. Macrophage diversity enhances tumor progression and metastasis. Cell. 2010;141(1):39–51. doi: 10.1016/j.cell.2010.03.014. [ DOI ] [ PMC free article ] [ PubMed ] [ Google Scholar ]
  • 52. Egeblad M, Ewald AJ, Askautrud HA, Truitt ML, Welm BE, Bainbridge E, Peeters G, Krummel MF, Werb Z. Visualizing stromal cell dynamics in different tumor microenvironments by spinning disk confocal microscopy. Dis Model Mech. 2008;1(2–3):155–167. doi: 10.1242/dmm.000596. discussion 165.. ••The authors utilize spinning disk confocal microscopy to examine behavior of various genetically labeled cell types in different tumor microenvironments of the same mouse, enabling functional analysis of disctinct cell population in the same tumors.
  • 53. Vakoc BJ, Lanning RM, Tyrrell JA, Padera TP, Bartlett LA, Stylianopoulos T, Munn LL, Tearney GJ, Fukumura D, Jain RK, Bouma BE. Three-dimensional microscopy of the tumor microenvironment in vivo using optical frequency domain imaging. Nat Med. 2009;15(10):1219–1223. doi: 10.1038/nm.1971. [ DOI ] [ PMC free article ] [ PubMed ] [ Google Scholar ]
  • 54. Carbonell WS, Ansorge O, Sibson N, Muschel R. The vascular basement membrane as "soil" in brain metastasis. PLoS ONE. 2009;4(6):e5857. doi: 10.1371/journal.pone.0005857. [ DOI ] [ PMC free article ] [ PubMed ] [ Google Scholar ]
  • 55. Kienast Y, von Baumgarten L, Fuhrmann M, Klinkert WE, Goldbrunner R, Herms J, Winkler F. Real-time imaging reveals the single steps of brain metastasis formation. Nat Med. 2010;16(1):116–122. doi: 10.1038/nm.2072.. ••Through the use of multiphoton laser scaning microscopy, the authors are able to track individual brain metastatic cell fate in relationship to the blood vessels over minutes to months, establishing essential steps in the process of brain metastasis and response to therapy.
  • 56. Olson ES, Aguilera TA, Jiang T, Ellies LG, Nguyen QT, Wong EH, Gross LA, Tsien RY. In vivo characterization of activatable cell penetrating peptides for targeting protease activity in cancer. Integr Biol (Camb) 2009;1(5–6):382–393. doi: 10.1039/b904890a. [ DOI ] [ PMC free article ] [ PubMed ] [ Google Scholar ]
  • 57. Macchiarini F, Manz MG, Palucka AK, Shultz LD. Humanized mice: are we there yet? J Exp Med. 2005;202(10):1307–1311. doi: 10.1084/jem.20051547. [ DOI ] [ PMC free article ] [ PubMed ] [ Google Scholar ]
  • 58. Wang J, Xia TS, Liu XA, Ding Q, Du Q, Yin H, Wang S. A novel orthotopic and metastatic mouse model of breast cancer in human mammary microenvironment. Breast Cancer Res Treat. 2010;120(2):337–344. doi: 10.1007/s10549-009-0391-4. [ DOI ] [ PubMed ] [ Google Scholar ]
  • 59. Kuperwasser C, Chavarria T, Wu M, Magrane G, Gray JW, Carey L, Richardson A, Weinberg RA. Reconstruction of functionally normal and malignant human breast tissues in mice. Proc Natl Acad Sci U S A. 2004;101(14):4966–4971. doi: 10.1073/pnas.0401064101. [ DOI ] [ PMC free article ] [ PubMed ] [ Google Scholar ]
  • 60. Moody SE, Sarkisian CJ, Hahn KT, Gunther EJ, Pickup S, Dugan KD, Innocent N, Cardiff RD, Schnall MD, Chodosh LA. Conditional activation of Neu in the mammary epithelium of transgenic mice results in reversible pulmonary metastasis. Cancer Cell. 2002;2(6):451–461. doi: 10.1016/s1535-6108(02)00212-x. [ DOI ] [ PubMed ] [ Google Scholar ]
  • 61. Podsypanina K, Politi K, Beverly LJ, Varmus HE. Oncogene cooperation in tumor maintenance and tumor recurrence in mouse mammary tumors induced by Myc and mutant Kras. Proc Natl Acad Sci U S A. 2008;105(13):5242–5247. doi: 10.1073/pnas.0801197105. [ DOI ] [ PMC free article ] [ PubMed ] [ Google Scholar ]
  • 62. Felsher DW, Bishop JM. Reversible tumorigenesis by MYC in hematopoietic lineages. Mol Cell. 1999;4(2):199–207. doi: 10.1016/s1097-2765(00)80367-6. [ DOI ] [ PubMed ] [ Google Scholar ]
  • 63. Stern P, Astrof S, Erkeland SJ, Schustak J, Sharp PA, Hynes RO. A system for Creregulated RNA interference in vivo. Proc Natl Acad Sci U S A. 2008;105(37):13895–13900. doi: 10.1073/pnas.0806907105. [ DOI ] [ PMC free article ] [ PubMed ] [ Google Scholar ]
  • 64. Dickins RA, McJunkin K, Hernando E, Premsrirut PK, Krizhanovsky V, Burgess DJ, Kim SY, Cordon-Cardo C, Zender L, Hannon GJ, Lowe SW. Tissue-specific and reversible RNA interference in transgenic mice. Nat Genet. 2007;39(7):914–921. doi: 10.1038/ng2045. [ DOI ] [ PMC free article ] [ PubMed ] [ Google Scholar ]
  • View on publisher site
  • PDF (526.3 KB)
  • Collections

Similar articles

Cited by other articles, links to ncbi databases.

  • Download .nbib .nbib
  • Format: AMA APA MLA NLM

Add to Collections

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • View all journals
  • Explore content
  • About the journal
  • Publish with us
  • Sign up for alerts
  • Published: 24 January 2011

Mouse models of advanced spontaneous metastasis for experimental therapeutics

  • Giulio Francia 1 ,
  • William Cruz-Munoz 1 ,
  • Shan Man 1 ,
  • Ping Xu 1 &
  • Robert S. Kerbel 1  

Nature Reviews Cancer volume  11 ,  pages 135–141 ( 2011 ) Cite this article

14k Accesses

286 Citations

7 Altmetric

Metrics details

  • Cancer therapy
  • Drug development

An enduring problem in cancer research is the failure to reproduce highly encouraging preclinical therapeutic findings using transplanted or spontaneous primary tumours in mice in clinical trials of patients with advanced metastatic disease. There are several reasons for this, including the failure to model established, visceral metastatic disease. We therefore developed various models of aggressive multi-organ spontaneous metastasis after surgical resection of orthotopically transplanted human tumour xenografts. In this Opinion article we provide a personal perspective summarizing the prospect of their increased clinical relevance. This includes the reduced efficacy of certain targeted anticancer drugs, the late emergence of spontaneous brain metastases and the clinical trial results evaluating a highly effective therapeutic strategy previously tested using such models.

This is a preview of subscription content, access via your institution

Access options

Subscribe to this journal

Receive 12 print issues and online access

$209.00 per year

only $17.42 per issue

Buy this article

  • Purchase on SpringerLink
  • Instant access to full article PDF

Prices may be subject to local taxes which are calculated during checkout

mouse experimental metastasis model

Similar content being viewed by others

mouse experimental metastasis model

Dissecting metastasis using preclinical models and methods

mouse experimental metastasis model

Experimental and spontaneous metastasis assays can result in divergence in clonal architecture

mouse experimental metastasis model

Emerging strategies for treating metastasis

Wilmanns, C., Fan, D., O'Brian, C. A., Bucana, C. D. & Fidler, I. J. Orthotopic and ectopic organ environments differentially influence the sensitivity of murine colon carcinoma cells to doxorubicin and 5-fluorouracil. Int. J. Cancer 52 , 98–104 (1992).

Article   CAS   Google Scholar  

Kerbel, R. S. What is the optimal rodent model for anti-tumor drug testing? Cancer Metastasis Rev. 17 , 301–304 (1998).

Article   Google Scholar  

Kerbel, R. S. Human tumor xenografts as predictive preclinical models for anticancer drug activity in humans: better than commonly perceived - but they can. be improved. Cancer Biol. Ther. 2 , 108–113 (2003).

Google Scholar  

Peterson, J. K. & Houghton, P. J. Integrating pharmacology and in vivo cancer models in preclinical and clinical drug development. Eur. J. Cancer 40 , 837–844 (2004).

Van Dyke, T. & Jacks, T. Cancer modeling in the modern era: progress and challenges. Cell 108 , 135–144 (2002).

Ottewell, P. D., Coleman, R. E. & Holen, I. From genetic abnormality to metastases: murine models of breast cancer and their use in the development of anticancer therapies. Breast Cancer Res. Treat. 96 , 101–113 (2006).

Talmadge, J. E., Singh, R. K., Fidler, I. J. & Raz, A. Murine models to evaluate novel and conventional therapeutic strategies for cancer. Am. J. Pathol. 170 , 793–804 (2007).

Brodie, S. G. et al. Multiple genetic changes are associated with mammary tumorigenesis in Brca1 conditional knockout mice. Oncogene 20 , 7514–7523 (2001).

Stambolic, V. et al. High incidence of breast and endometrial neoplasia resembling human Cowden syndrome in pten± mice. Cancer Res. 60 , 3605–3611 (2000).

CAS   PubMed   Google Scholar  

Gourley, C. et al. Increased incidence of visceral metastases in scottish patients with BRCA1/2-defective ovarian cancer: an extension of the ovarian BRCAness phenotype. J. Clin. Oncol. 28 , 2505–2511 (2010).

Albiges, L. et al. Spectrum of breast cancer metastasis in BRCA1 mutation carriers: highly increased incidence of brain metastases. Ann. Oncol. 16 , 1846–1847 (2005).

Schmitz, M. et al. Complete loss of PTEN expression as a possible early prognostic marker for prostate cancer metastasis. Int. J. Cancer 120 , 1284–1292 (2007).

Khanna, C. & Hunter, K. Modeling metastasis in vivo . Carcinogenesis 26 , 513–523 (2005).

Sharpless, N. E. & DePinho, R. A. The mighty mouse: genetically engineered mouse models in cancer drug development. Nature Rev. Drug Discov. 5 , 741–754 (2006).

Heyer, J., Kwong, L. N., Lowe, S. W. & Chin, L. Non-germline genetically engineered mouse models for translational cancer research. Nature Rev. Cancer 10 , 470–480 (2010).

Kim, I. S. & Baek, S. H. Mouse models for breast cancer metastasis. Biochem. Biophys. Res. Commun. 394 , 443–447 (2010).

Paez-Ribes, M. et al. Antiangiogenic therapy elicits malignant progression of tumors to increased local invasion and distant metastasis. Cancer Cell 15 , 220–231 (2009).

Bergers, G., Song, S., Meyer-Morse, N., Bergsland, E. & Hanahan, D. Benefits of targeting both pericytes and endothelial cells in the tumor vasculature with kinase inhibitors. J. Clin. Invest. 111 , 1287–1295 (2003).

Singh, M. et al. Assessing therapeutic responses in Kras mutant cancers using genetically engineered mouse models. Nature Biotech. 28 , 585–593 (2010).

Francia, G. & Kerbel, R. S. Raising the bar for cancer therapy models. Nature Biotech. 28 , 561–562 (2010).

Van, D. T. Approximating a human cancer. Nature Med. 16 , 976–977 (2010).

Olive, K. P. et al. Inhibition of Hedgehog signaling enhances delivery of chemotherapy in a mouse model of pancreatic cancer. Science 324 , 1457–1461 (2009).

Morikawa, K., Walker, S. M., Jessup, J. M. & Fidler, I. J. In vivo selection of highly metastatic cells from surgical specimens of different primary human colon carcinomas implanted into nude mice. Cancer Res. 48 , 1943–1948 (1988).

Kubota, T. Metastatic models of human cancer xenografted in the nude mouse: the importance of orthotopic transplantation. J. Cell. Biochem. 56 , 4–8 (1994).

Furukawa, T., Kubota, T., Watanabe, M., Kitajima, M. & Hoffman, R. M. A novel “patient-like” treatment model of human pancreatic cancer constructed using orthotopic transplantation of histologically intact human tumor tissue in nude mice. Cancer Res. 53 , 3070–3072 (1993).

Kiguchi, K. et al. A patient-like orthotopic implantation nude mouse model of highly metastatic human ovarian cancer. Clin. Exp. Metastasis 16 , 751–756 (1998).

Price, J. E. & Zhang, R. D. Studies of human breast cancer metastasis using nude mice. Cancer Metastasis Rev. 8 , 285–297 (1990).

Fidler, I. J. Models for spontaneous metastasis. Cancer Res. 66 , 9787 (2006).

Dawson, M. R., Duda, D. G., Fukumura, D. & Jain, R. K. VEGFR1-activity-independent metastasis formation. Nature 461 , e4 (2009).

Nangia-Makker, P. et al. Inhibition of human cancer cell growth and metastasis in nude mice by oral intake of modified citrus pectin. J. Natl Cancer Inst. 94 , 1854–1862 (2002).

Teicher, B. A. et al. Potentiation of cytotoxic cancer therapies by TNP-470 alone and with other anti-angiogenic agents. Int. J. Cancer 57 , 920–925 (1994).

Holden, S. A., Emi, Y., Kakeji, Y., Northey, D. & Teicher, B. A. Host distribution and response to antitumor alkylating agents of EMT-6 tumor cells from subcutaneous tumor implants. Cancer Chemother. Pharmacol. 40 , 87–93 (1997).

Barnett, S. C. & Eccles, S. A. Studies of mammary carcinoma metastasis in a mouse model system. II: lectin binding properties of cells in relation to the incidence and organ distribution of metastases. Clin. Exp. Metastasis 2 , 297–310 (1984).

Munoz, R. et al. Highly efficacious non-toxic treatment for advanced metastatic breast cancer using combination UFT-cyclophosphamide metronomic chemotherapy. Cancer Res. 66 , 3386–3391 (2006).

Cruz-Munoz, W., Man, S., Xu, P. & Kerbel, R. S. Development of a preclinical model of spontaneous human melanoma CNS metastasis. Cancer Res. 68 , 4500–4505 (2008).

Francia, G. et al. Long term progression and therapeutic response of visceral metastatic disease non-invasively monitored in mouse urine using β-hCG choriogonadotropin secreting tumor cell lines. Mol. Cancer Ther. 7 , 3452–3459 (2008).

Fidler, I. J. & Hart, I. R. Biological diversity in metastatic neoplasms: origins and implications. Science 217 , 998–1003 (1982).

Barnett, S. C. & Eccles, S. A. Studies of mammary carcinoma metastasis in a mouse model system. I: derivation and characterization of cells with different metastatic properties during tumour progression in vivo . Clin. Exp. Metastasis 2 , 15–36 (1984).

Shih, I. M. et al. Assessing tumors in living animals through measurement of urinary β-human chorionic gonadotropin. Nature Med. 6 , 711–714 (2000).

Hoffman, R. M. Imaging cancer dynamics in vivo at the tumor and cellular level with fluorescent proteins. Clin. Exp. Metastasis 26 , 345–355 (2009).

Sahai, E. Illuminating the metastatic process. Nature Rev. Cancer 7 , 737–749 (2007).

Kozlowski, J. M. et al. Metastatic behavior of human tumor cell lines grown in the nude mouse. Cancer Res. 44 , 3522–3529 (1984).

Poste, G., Doll, J., Hart, I. R. & Fidler, I. J. In vitro selection of murine B16 melanoma variants with enhanced tissue-invasive properties. Cancer Res. 40 , 1636–1644 (1980).

Kerbel, R. S., Man, M. S. & Dexter, D. A model of human cancer metastasis: extensive spontaneous and artificial metastasis of a human pigmented and derived variant sublines in nude mice. J. Natl Cancer Inst. 72 , 93–108 (1984).

Price, J. E., Polyzos, A., Zhang, R. D. & Daniels, L. M. Tumorigenicity and metastasis of human breast carcinoma cell lines in nude mice. Cancer Res. 50 , 717–721 (1990).

Francia, G. et al. Comparative impact of trastuzumab and cyclophosphamide on HER-2 positive human breast cancer xenografts. Clin. Cancer Res. 15 , 6358–6366 (2009).

du Manoir, J. M. et al. Strategies for delaying or treating in vivo acquired resistance to trastuzumab (Herceptin ® ) in human breast cancer xenografts. Clin. Cancer Res. 12 , 904–916 (2006).

Man, S. et al. Antitumor and anti-angiogenic effects in mice of low-dose (metronomic) cyclophosphamide administered continuously through the drinking water. Cancer Res. 62 , 2731–2735 (2002).

Hurwitz, H. et al. Bevacizumab plus irinotecan, fluorouracil, and leucovorin for metastatic colorectal cancer. N. Engl. J. Med. 350 , 2335–2342 (2004).

Hudis, C. A. Trastuzumab-mechanism of action and use in clinical practice. N. Engl. J. Med. 357 , 39–51 (2007).

Kerbel, R. S. Antiangiogenic therapy: a universal chemosensitization strategy for cancer? Science 312 , 1171–1175 (2006).

Burger, R. A. et al. Phase III trial of bevacizumab (BEV) in the primary treatment of advanced epithelial ovarian cancer (EOC), primary peritoneal cancer (PPC), or fallopian tube cancer (FTC): a Gynecologic Oncology Group study. J. Clin. Oncol. Abst. 28 , LBA1 (2010).

Man, S., Munoz, R. & Kerbel, R. S. On the development of models in mice of advanced visceral metastatic disease for anti-cancer drug testing. Cancer Metastasis Rev. 26 , 737–747 (2007).

Gril, B. et al. Effect of lapatinib on the outgrowth of metastatic breast cancer cells to the brain. J. Natl Cancer Inst. 100 , 1092–1103 (2008).

Warren, R. S., Yuan, H., Mati, M. R., Gillett, N. A. & Ferrara, N. Regulation by vascular endothelial growth factor of human colon cancer tumorigenesis in a mouse model of experimental liver metastasis. J. Clin. Invest. 95 , 1789–1797 (1995).

Vantyghem, S. A. et al. Dietary genistein reduces metastasis in a postsurgical orthotopic breast cancer model. Cancer Res. 65 , 3396–3403 (2005).

McCulloch, P. & George, W. D. Warfarin inhibits metastasis of Mtln3 rat mammary carcinoma without affecting primary tumour growth. Br. J. Cancer 59 , 179–183 (1989).

Dellapasqua, S. et al. Metronomic cyclophosphamide and capecitabine combined with bevacizumab in advanced breast cancer: clinical and biological activity. J. Clin. Oncol. 26 , 4899–4905 (2008).

Cruz-Munoz, W., Man, S. & Kerbel, R. S. Effective treatment of advanced human melanoma metastasis in immunodeficient mice using combination metronomic chemotherapy regimens. Clin. Cancer Res. 15 , 4867–4874 (2009).

Lawson, D. H. Choices in adjuvant therapy of melanoma. Cancer Control 12 , 236–241 (2005).

Lin, N. U. & Winer, E. P. Brain metastases: the HER2 paradigm. Clin. Cancer Res. 13 , 1648–1655 (2007).

Steeg, P. S. et al. Preclinical drug development must consider the impact on metastasis. Clin. Cancer Res. 15 , 4529–4530 (2009).

Palmieri, D. et al. Vorinostat inhibits brain metastatic colonization in a model of triple-negative breast cancer and induces DNA double-strand breaks. Clin. Cancer Res. 15 , 6148–6157 (2009).

Carbonell, W. S., Ansorge, O., Sibson, N. & Muschel, R. The vascular basement membrane as “soil” in brain metastasis. PLoS ONE 4 , e5857 (2009).

Minn., A. J. et al. Distinct organ-specific metastatic potential of individual breast cancer cells and primary tumors. J. Clin. Invest. 115 , 44–55 (2005).

Zhang, X. H. et al. Latent bone metastasis in breast cancer tied to Src-dependent survival signals. Cancer Cell 16 , 67–78 (2009).

Welch, D. R. Technical considerations for studying cancer metastasis in vivo . Clin. Exp. Metastasis 15 , 272–306 (1997).

Steeg, P. S. & Theodorescu, D. Metastasis: a therapeutic target for cancer. Nature Clin. Pract. Oncol. 5 , 206–219 (2008).

Dunn, L. K. et al. Hypoxia and TGF-β drive breast cancer bone metastases through parallel signaling pathways in tumor cells and the bone microenvironment. PLoS ONE 4 , e6896 (2009).

Kang, Y. et al. A multigenic program mediating breast cancer metastasis to bone. Cancer Cell 3 , 537–549 (2003).

Bos, P. D. et al. Genes that mediate breast cancer metastasis to the brain. Nature 459 , 1005–1009 (2009).

Minn, A. J. et al. Genes that mediate breast cancer metastasis to lung. Nature 436 , 518–524 (2005).

Price, J. E. Analyzing the metastatic phenotype. J. Cell. Biochem. 56 , 16–22 (1994).

Download references

Acknowledgements

The authors would like to thank C. Cheng for her excellent secretarial assistance; L. Ellis, I. Hart, I. J. Fidler, C. Hackl and U. Emmenegger for their comments, past and present. R.S.K.'s metastasis therapy studies are supported by grants from the US National Institutes of Health (NIH)(CA-41233), Canadian Cancer Society Research Institute (CCSRI), Canadian Institutes of Health Research (CIHR) and the Ontario Institute for Cancer Research (OICR), as well as past or present sponsored research agreements with Taiho Pharmaceuticals, Tokyo, Japan, ImClone Systems, New York, GSK, Philadelphia and Pfizer, La Jolla, USA. R.S.K. holds a Tier I Canada Research Chair in Tumour Biology, Angiogenesis and Antiangiogenic Therapy.

Author information

Authors and affiliations.

Giulio Francia, William Cruz-Munoz, Shan Man, Ping Xu and Robert S. Kerbel are at The Molecular & Cellular Biology Research, Sunnybrook Health Science Centre, S-217 Research Building, 2075 Bayview Avenue, Toronto, Ontario M4N 3M5, Canada.,

Giulio Francia, William Cruz-Munoz, Shan Man, Ping Xu & Robert S. Kerbel

You can also search for this author in PubMed   Google Scholar

Corresponding authors

Correspondence to Giulio Francia or Robert S. Kerbel .

Ethics declarations

Competing interests.

R.S.K. is a consultant at Taiho Pharmaceuticals, Japan, and a member of the Scientific Advisory Board at MetronomX, USA.

Related links

Further information.

Robert S. Kerbel's homepage

Clinicaltrails.gov Identifier NCT01131195

Rights and permissions

Reprints and permissions

About this article

Cite this article.

Francia, G., Cruz-Munoz, W., Man, S. et al. Mouse models of advanced spontaneous metastasis for experimental therapeutics. Nat Rev Cancer 11 , 135–141 (2011). https://doi.org/10.1038/nrc3001

Download citation

Published : 24 January 2011

Issue Date : February 2011

DOI : https://doi.org/10.1038/nrc3001

Share this article

Anyone you share the following link with will be able to read this content:

Sorry, a shareable link is not currently available for this article.

Provided by the Springer Nature SharedIt content-sharing initiative

This article is cited by

Role of gut microbiota in predisposition to colon cancer: a narrative review.

  • Hattiangadi Shruthi Kamath
  • Rushikesh Shukla
  • Dibyankita Ghosh

Indian Journal of Microbiology (2024)

Current landscape and challenges ahead of immuno-molecular mechanism and immunotherapy strategy of brain metastases

Holistic Integrative Oncology (2023)

The pro-tumorigenic responses in metastatic niches: an immunological perspective

  • Mohammad Hossein Mirzapour
  • Mahsa Heidari-Foroozan
  • Nima Rezaei

Clinical and Translational Oncology (2022)

Impact of limb amputation and cisplatin chemotherapy on metastatic progression in mouse models of osteosarcoma

  • Amy K. LeBlanc

Scientific Reports (2021)

A common goal to CARE: Cancer Advocates, Researchers, and Clinicians Explore current treatments and clinical trials for breast cancer brain metastases

  • Natalie S. Joe
  • Christine Hodgdon
  • Daniele M. Gilkes

npj Breast Cancer (2021)

Quick links

  • Explore articles by subject
  • Guide to authors
  • Editorial policies

Sign up for the Nature Briefing: Cancer newsletter — what matters in cancer research, free to your inbox weekly.

mouse experimental metastasis model

An official website of the United States government

Official websites use .gov A .gov website belongs to an official government organization in the United States.

Secure .gov websites use HTTPS A lock ( Lock Locked padlock icon ) or https:// means you've safely connected to the .gov website. Share sensitive information only on official, secure websites.

  • Publications
  • Account settings
  • Advanced Search
  • Journal List

Chinese Medical Journal logo

Experimental research on spinal metastasis with mouse models

Xiaochen qiao, zelong song, zhuohao liu, xuesong zhang, xiangyu wang.

  • Author information
  • Article notes
  • Copyright and License information

Correspondence to: Xiangyu Wang, Department of Pain Medicine, First Medical Center, the PLA General Hospital, Beijing 100853, ChinaE-Mail: [email protected]

Received 2023 Mar 11; Issue date 2023 Dec 20.

This is an open access article distributed under the terms of the Creative Commons Attribution-Non Commercial-No Derivatives License 4.0 (CCBY-NC-ND), where it is permissible to download and share the work provided it is properly cited. The work cannot be changed in any way or used commercially without permission from the journal. http://creativecommons.org/licenses/by-nc-nd/4.0

To the Editor : The spine is a common site for tumor metastasis. Patients with spinal metastases have pain, pathological fractures and spinal deformities owing to the tumor invasion of spinal bones. At present, the mouse model is a very important tool for spinal metastases-related studies. [ 1 ] It is crucial to elucidate the pathophysiological mechanism underlying spinal metastases, improve the diagnosis and treatment guidelines, and develop new therapeutic methods. In this study, we have summarized various mouse spinal metastasis models and the applicable research contents.

Presently, human and mouse tumor cell lines have been used to produce mouse spinal metastatic tumor models. Human tumor cell lines include prostate cancer PC-3, a series of lung cancer cell lines (PC-9, A549, NCI-H1299, NCI-H460, H2030, SPCA-1, and PC-14), melanoma A2058, kidney cancer ACHN, and breast cancer. Murine derived tumor cell lines include BALB/c mouse breast cancer TM40D, breast cancer 4T1, melanoma B16, melanoma B16-luc, melanoma mB16-luc, melanoma B16-F1, prostate cancer MBT-2, prostate cancer TRAMP-C2, and lung cancer LLC1. As the murine tumor cell lines have the advantage of avoiding excessive host versus graft reaction [ 1 ] , they can be used to study the pathophysiological procedure and the molecular mechanisms at various stages of primary tumor metastasis and colonization in the spine. On the contrary, immunodeficient mice retain some of their immune function when human tumor cells are inoculated into immunodeficient mice, and the immune system of the mice will have a certain degree of host versus graft reaction, which can adversely affect the interpretation of tumor-related studies. However, because of the existence of species differences between mouse and human, spinal metastatic tumor models using murine tumor cells are not completely suitable for developing new therapies for human metastatic tumors. [ 2 ] Therefore, models of spinal metastases using human-derived mice are essential.

Some models using murine-derived tumor cells were established using non-immunodeficient mice, including BALB/C mice, C57BL/6J and C3H/He. In the spinal metastatic tumor model using human tumor cells, the carrier mice are immunodeficient, among which the T lymphocyte dysfunction mice are the mostly used. Moreover, some models used severe combined immunodeficiency non-obese diabetic/severe combined immunodeficiency (NOD/SCID) mice as carrier animals. The success rate of tumor modeling in these mice is higher, however, NOD/SCID mice are raised in more demanding conditions and are more prone to disease and death. [ 1 ]

The existing modeling methods include the circulatory system injection, the spine injection, and spontaneous spine metastasis. In the circulatory system injection method, a certain amount of tumor cell suspension is injected into the blood circulation system of mice to simulate the spread of tumor cells to the spine. In the spine injection method, the tumor mass or tumor cells are directly injected into the spine through surgery or percutaneous injection. In the spontaneous spine metastasis method, the tumor cell suspension is implanted in the corresponding in situ organ or subcutaneous tumors are implanted in mice and the cancer cells spontaneously metastasized in the spine to develop a spinal metastatic tumor after some time. [ 1 ]

In vivo bioluminescent imaging (BLI), micro-computed tomography (micro CT), magnetic resonance imaging (MRI), and positron emission tomography-CT (PET-CT) have been widely used to evaluate and monitor spinal metastasis mouse models. BLI can evaluate whether tumor tissues or cells inoculated are active immediately after injection, timely detect the existence of metastatic tumors in the initial stage of spinal metastases in mice before bone damage occurs, and timely record the occurrence time of metastatic tumors. Micro CT, which is used to evaluate osteoblastic or osteolytic changes in spinal metastases, can quantitatively analyze the bone volume, density and surface area and reconstruct the 3-dimensional image of bone tumors. MRI can perform multiple imaging tests on all soft tissues with high sensitivity. [ 3 ] Due to the limited resolution of MRI and micro CT, the presence of metastatic tumors can only be detected when the vertebral structure of mice is significantly changed by spinal metastases. Because of the increase of tracer uptake levels in tumor lesions before bone destruction, PET-CT using fluorodeoxyglucose has better sensitivity and accuracy but worse prediction for detecting spinal metastases in mice than MRI, indicating that it can detect smaller tumor tissues earlier. [ 4 ]

Extensive spinal metastases can compress the spinal cord, which leads to neurological dysfunction. Therefore, neurological dysfunction in a mouse model of metastatic spinal tumors is also the focus of research. Current studies focus on neuromotor and sphincter dysfunction evaluation. The most commonly neuromotor function evaluation method in studies is the Basso Mouse Scale (BMS) for Locomotion, [ 5 ] which formulated the characteristics of motor function changes in mice with spinal cord injury. Inconsistencies in lower limb functions that may be caused by spinal cord injury are considered, making it more perfect. The scale is sensitive, reliable, and effective. Instead of the BMS, researchers proposed classifying motor nerve function injury caused by tumor compression in mice into four landmark stages: tail dragging, dorsal stepping, hind-limb sweeping, and paralysis. In addition, some researchers performed gait analysis to evaluate neurological functions in mice with spinal metastases, which can provide a more objective evaluation of motor functions in mice. [ 1 ]

In this study, we have provided an overview of existing mouse models of spinal metastasis and explored the applicability and shortcomings of each modeling method. Spontaneous spinal metastasis can completely simulate the process of primary tumor metastasis to the spine. It is possible to study each step of the multi-step cascade of tumor metastasis, including the formation of a microenvironment before metastasis, the organ-targeting of tumor metastasis to the spine, the dormancy of tumor cells, the effect of surgical resection of primary tumors on metastasis, and the effect and regulatory function of the immune system on the formation of spinal metastasis. [ 1 ] Studies using the circulatory injection method mostly focus on the key steps of the early formation of spinal metastases, including the mechanisms of tumor cell propagation, circulation, and extravasation, with low complexity and difficulty in operation. However, this method it cannot model specific vertebral segments and can only randomly generate metastatic tumors on multiple different vertebral bodies, and this modeling method always resulted in tumors in multiple organs of the mice, which induced mice dying of infection and excessive tumor load before they develop neurological damage caused by spinal metastatic compression. Moreover, the timing, location, and number of spinal metastases in mice injected with tumor cells can vary and are unpredictable. [ 1 ] Transplanting tumors directly into the spine can produce highly consistent, reproducible and stable spinal metastatic tumor models. Researchers can use this modeling method to study the microenvironment of spinal metastases, the effects of metastatic tumors on the spinal peripheral nervous system, mechanisms related to metastatic tumor growth in the spine, the invasion of spinal bone tissues, the differences in the biological behavior of different tumor cell types in the spine, and the neurological dysfunction caused by spinal cord compression due to tumors. However, this method cannot be used to study tumor metastasis mechanisms. [ 1 ] The pathways of primary tumor metastasis to the spine include hematogenous spread, direct extension, and cerebrospinal fluid (CSF) spread. “Hematogenous spread” means that tumor cells metastasize from the primary tumor to the spine through blood flow. “Direct extension” means that primary tumors in soft tissues adjacent to vertebrae directly invade and spread to the spine. “CSF spread” means that brain tumors can metastasize to the spine via CSF. No mouse model of spinal metastasis is available that simulates the pathways of direct extension and CSF spread. Therefore, new mouse models of spinal metastasis need to be developed to simulate the process of spinal metastasis generation.

This work is supported by a grant from the Natural Science Foundation of Shanxi Province (No. 202204041101023).

Conflicts of interest

Kun Zhang, Yi Feng, Xiaochen Qiao contributed equally to this work.

How to cite this article: Zhang K, Feng Y, Qiao XC, Yu Y, Song ZL, Liu ZH, Tian Z, Chen S, Zhang XS, Wang XY. Experimental research on spinal metastasis with mouse models. Chin Med J 2023;136:3008–3009. doi: 10.1097/CM9.0000000000002922

  • 1. Jelgersma C, Vajkoczy P. How to Target Spinal Metastasis in Experimental Research: An Overview of Currently Used Experimental Mouse Models and Future Prospects. Int J Mol Sci 2021;22:5420. doi: 10.3390/ijms22115420 [ DOI ] [ PMC free article ] [ PubMed ] [ Google Scholar ]
  • 2. Tatsui CE, Lang FF, Gumin J, Suki D, Shinojima N, Rhines LD. An orthotopic murine model of human spinal metastasis: histological and functional correlations. J Neurosurg Spine 2009;10:501–512. doi: 10.3171/2009.2.SPINE08391. [ DOI ] [ PMC free article ] [ PubMed ] [ Google Scholar ]
  • 3. Hibberd C, Cossigny DA, Quan GM. Animal cancer models of skeletal metastasis. Cancer Growth Metastasis 2013;6:23–34. doi: 10.4137/CGM.S11284. [ DOI ] [ PMC free article ] [ PubMed ] [ Google Scholar ]
  • 4. Hu W, Liu Z, Xiao X, Yang Y, Sun Z, Wang X. Comparison of diagnostic efficacy of MRI and PET/CT in lung cancer of mouse with spinal metastasis. Cell Mol Biol 2020;66:138–142. [ PubMed ] [ Google Scholar ]
  • 5. Basso DM, Fisher LC, Anderson AJ, Jakeman LB, McTigue DM, Popovich PG. Basso Mouse Scale for locomotion detects differences in recovery after spinal cord injury in five common mouse strains. J Neurotrauma 2006;23:635–659. doi: 10.1089/neu.2006.23.635. [ DOI ] [ PubMed ] [ Google Scholar ]
  • View on publisher site
  • PDF (120.4 KB)
  • Collections

Similar articles

Cited by other articles, links to ncbi databases.

  • Download .nbib .nbib
  • Format: AMA APA MLA NLM

Add to Collections

IMAGES

  1. Characterizing in vivo imageable melanoma brain metastasis mouse...

    mouse experimental metastasis model

  2. Different tumorigenicity and distinct metastasis and gene signature between orthotopic and

    mouse experimental metastasis model

  3. Mouse model for breast cancer metastasis to brain and therapeutic...

    mouse experimental metastasis model

  4. Modeling metastasis in mice: a closer look: Trends in Cancer

    mouse experimental metastasis model

  5. Mouse Model of Cancer Metastasis

    mouse experimental metastasis model

  6. Cancers

    mouse experimental metastasis model

COMMENTS

  1. Experimental models for cancer brain metastasis - ScienceDirect

    Jan 1, 2024 · The first mouse metastasis model was the B16 melanoma model, which was developed from a spontaneous mouse tumor that metastasized to the lung and brain when reintroduced into syngeneic mice. 31 The B16 model laid the foundation for the seminal work of Fidler et al., which established many fundamental concepts of metastasis progression ...

  2. Mouse models of advanced spontaneous metastasis for ...

    Furthermore, the use of experimental metastasis assays has also been highly successful for investigating the tissue specificity of metastases (for example, for experimental breast cancer metastases 65,66,70, using models that are also capable of spontaneous metastasis 71,72), as well as their response to targeted therapies 55 and the ...

  3. Mouse Models for Tumor Metastasis - PMC - PubMed Central (PMC)

    The second model is an experimental metastasis model. In this model, luciferase-labeled MDA-MB-231 human breast tumor cells are injected into the tail vein of NOD-SCID immunodeficient mice and the presence of tumor cells in mice will be detected through the noninvasive bioluminescence imaging ( 13 , 14 ).

  4. Modeling metastasis in the mouse - PMC

    Clinically, metastasis may take decades to become manifest in certain types of cancer , and there is a dearth of mouse models to study the biology of the latent metastatic state . Both of these questions would benefit from the development of additional immunocompromised models (rev. in [ 57 ]).

  5. Mouse models of advanced spontaneous metastasis for ... - Nature

    Jan 24, 2011 · Barnett, S. C. & Eccles, S. A. Studies of mammary carcinoma metastasis in a mouse model system. II: lectin binding properties of cells in relation to the incidence and organ distribution of ...

  6. Mouse models of metastasis: progress and prospects

    Sep 1, 2017 · More recently, elegant mouse experiments again using experimental metastasis models have shown that tumor-derived exosomes (see Glossary, Box 1) induce pro-metastatic progenitor cells in the bone marrow through receptor tyrosine kinase MET signaling (Peinado et al., 2012), and that exosomal integrins (see Glossary, Box 1) can direct organ ...

  7. Modeling metastasis in the mouse - ScienceDirect

    Oct 1, 2010 · Clinically, metastasis may take decades to become manifest in certain types of cancer [4], and there is a dearth of mouse models to study the biology of the latent metastatic state [2 ••]. Both of these questions would benefit from the development of additional immunocompromised models (reviewed in [ 57 ]).

  8. Mouse models of spontaneous liver and lung metastasis for ...

    Mar 15, 2024 · Metastasis is a multistep process beginning with intravasation. In this step, cancer cells disseminate from their primary site into the blood stream. 2 In a consecutive step, cancer cells extravasate from the bloodstream into other organs, adapt to their new microenvironment, and grow to form metastasis. 3 Depending on the location of the primary tumor, different metastatic sites can be ...

  9. Experimental models for cancer brain metastasis

    Oct 30, 2023 · The first mouse metastasis model was the B16 melanoma model, which was developed from a spontaneous mouse tumor that metastasized to the lung and brain when reintroduced into syngeneic mice.31 The B16 model laid the foundation for the seminal work of Fidler et al., which established many fundamental concepts of metastasis progression, including ...

  10. Experimental research on spinal metastasis with mouse models

    Dec 12, 2023 · Therefore, new mouse models of spinal metastasis need to be developed to simulate the process of spinal metastasis generation. Funding This work is supported by a grant from the Natural Science Foundation of Shanxi Province (No. 202204041101023).