You are on page 1of 7

Cancer Letters 387 (2017) 114–120

Contents lists available at ScienceDirect

Cancer Letters
j o u r n a l h o m e p a g e : w w w. e l s e v i e r. c o m / l o c a t e / c a n l e t

Mini-review

Animal models of colorectal cancer with liver metastasis


Bo Young Oh a, Hye Kyung Hong b, Woo Yong Lee a,c, Yong Beom Cho a,c,d,*
a Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
b Samsung Biomedical Research Institute, Samsung Medical Center, Sungkyunkwan University, Seoul, Republic of Korea
c Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul, Republic of Korea
d
Department of Medical Device Management & Research, SAIHST, Sungkyunkwan University, Seoul, Republic of Korea

A R T I C L E I N F O A B S T R A C T

Keywords:
Liver metastasis is a leading cause of death in patients with colorectal cancer. Investigating the mecha-
Colorectal cancer
nisms of liver metastasis and control of disease progression are important strategies for improving survival
Liver metastasis
Mouse model
of these patients. Liver metastasis is a multi-step process and relevant models representing these steps
Orthotopic model are necessary to understand the mechanism of liver metastasis and establish appropriate treatments.
Genetically engineered mouse model Recently, the development of animal models for use in metastasis research has greatly increased; however,
Xenografts there is still a lack of models that sufficiently represent human cancer. Thus, in order to select an optimal
model for of a given study, it is necessary to fully understand the characteristics of each animal model.
In this review, we describe the mouse models currently used for colorectal cancer with liver metastasis,
their characteristics, and their pros and cons. This may help us specify the mechanism of liver metas-
tasis and provide evidence relevant to clinical applications.
© 2016 Elsevier Ireland Ltd. All rights reserved.

Introduction Liver metastasis models have been developed both in vitro and
in vivo. In vitro metastasis models are easy to manipulate, inexpen-
Colorectal cancer is the third most common malignancy and a sive, and reproducible. These models allow for the manipulation of
leading cause of cancer-related death worldwide [1–4]. The sur- each step in metastasis, but there are limitations with respect to a
vival of patients with colorectal cancer has improved; however, the comprehensive analysis of the whole process of metastasis [13,14].
mortality rate still ranks third due to metastasis or recurrence [4,5]. In contrast, in vivo metastasis models using animals may more ac-
Metastases are found in about 20–25% of patients with colorectal curately represent the metastatic process and can be genetically
cancer at diagnosis, and will develop in about 50% of patients during manipulated to mimic human cancer. Thus, animal models have been
the course of the disease [6,7]. The liver is the most common site widely used in metastasis research. However, they are associated
of metastasis for colorectal cancer, and only 15–20% of patients with with ethical concerns and cost burdens [15–17]. Currently, a number
liver metastases are suitable candidates for surgical resection [8–10]. of animal models have been developed for the evaluation of the
Several adjunctive therapies have been applied in patients with mechanism of liver metastasis and for the establishment of ther-
unresectable liver metastasis, but survival rates remain unfavor- apeutic strategies [12,15–19]. Such preclinical studies have provided
able [10]. Thus, investigating the mechanisms of liver metastasis and evidence relevant to clinical applications and have contributed to
control of disease progression are important strategies for improv- improving the survival of patients with colorectal cancer with liver
ing survival of these patients. Metastasis is a multistep process metastasis. In this review, we focus on the mouse models cur-
which involves an epithelial–mesenchymal transition, suppres- rently used for colorectal cancer with liver metastasis.
sion of apoptosis, local invasion and cell migration, angiogenesis and
intravasation, and extravasation into distant organs [11]. During these Overview of the current understanding of animal models
processes, tumor cells continuously interact with the microenvi-
ronment of the host cell [12]. Relevant models representing the Ideal animal models need to satisfy several conditions in order
metastatic characteristics of tumor cells are necessary in under- to be suitable for metastasis research. Animal models of metasta-
standing these metastatic processes. In particular, organ-specific sis should allow for optimal tumor take of both primary and
metastasis models can help us to specify the mechanism of me- metastatic tumors. The tumor take should be predictable and re-
tastasis and, accordingly, establish appropriate treatments. producible. They should recapitulate the entire process of human
metastatic diseases. Furthermore, they should reflect the genetic al-
terations with subsequent changes occurring in the metastatic
* Corresponding author. Tel.: +82 2 3410 0217; fax: +82 2 3410 1655. process. Ultimately, they should be available to test and validate novel
E-mail address: gscyb@skku.edu (Y.B. Cho). therapeutics for cancer patients [14,19–21]. However, as no model

http://dx.doi.org/10.1016/j.canlet.2016.01.048
0304-3835/© 2016 Elsevier Ireland Ltd. All rights reserved.
B.Y. Oh et al. / Cancer Letters 387 (2017) 114–120 115

meets all these conditions, models should be chosen according to models have received attention with the rise of personalized med-
the specific experimental purpose. icine. PDX models can reflect the characteristics of each patient and
Several animal models have been used in metastasis research, are useful for predicting response to new therapeutics [16,33].
with mouse models being most commonly used. Mouse models have
potential advantages such as anatomical and biological similari- Types of mouse models for colorectal cancer with
ties to humans, small size, ease of handling, breeding capacity, short liver metastasis
gestation time, cost-effectiveness, and ease of genetic manipula-
tion [14,22–24]. Hence, mouse models have evolved according to Spontaneous liver metastasis models
the specific experimental purpose required to study the biology of
human cancer metastasis. Most metastasis models have been ob- There are several ways to establish liver metastasis models (Fig. 1).
tained by transplanting cancer cell lines or tissues into mice. The The first approach for developing liver metastasis is the implanta-
most commonly used types of mice in transplantable models are tion of cancer cell lines or tissues into the colon (orthotopic
nude mice and severely compromised immunodeficient (SCID) mice. transplantation) or subcutaneous layer (ectopic transplantation),
Nude mice have a mutation of the Foxn1 gene leading to an athymic which results in the formation of primary tumors at the injection
state. Thus, they exhibit depletion of T cells and impaired T and B site, followed by the spontaneous development of liver metasta-
cell function. SCID mice have a mutation of the Prkdc gene, result- sis. The most commonly used cell lines are HCT116 and HT29; other
ing in deficiencies in the number and function of both T and B cells. cell lines used include SW480, SW620, and Lovo [12,18,34–37].
However, they retain intact innate immune system components such Ectopic transplantation is the easiest method for tumor engraft-
as natural killer (NK) cells and macrophages [14,19,25–27]. In ad- ment. However, this model mostly fails to develop liver metastasis
dition, SCID-beige mice, non-obese diabetic (NOD)-SCID mice, NOD/ because the subcutaneous microenvironment is quite different from
Shi-scid IL2rγnull (NOG) or NOD-SCID gamma (NSG) mice, and that of the colon [38–40]. In contrast, the orthotopic transplanta-
recombination activating genes (RAG) mice have been used as mouse tion model is similar to human cancer in terms of histology,
models. These mice have different genomic mutations leading to vascularity, gene expression, and the metastatic process [41–43].
various levels of immunodeficiency. Genetically engineered mice Thus, this model is mainly used in spontaneous liver metastasis
(GEM), which, unlike transplantable models, are generated through models. In this model, colon cancer cell lines or human tumor tissues
alterations in the expression of genes of interest, have also been de- can be transplanted into the colonic wall (usually the cecum) of the
veloped. In these models, organ-specific oncogenes or tumor immuno-compromised mice by subserosal injection or surgical im-
suppressor genes are manipulated, leading to the spontaneous de- plantation (Fig. 2A and B) [14,19,27].
velopment of cancer [14,16,27,28]. To ensure the effective development of liver metastasis, in vivo
Transplantable mouse models are divided into syngeneic and xe- selection of highly metastatic tumor cells is an important strate-
nograft models. Syngeneic models refer to models developed through gy. The metastatic potential of the engrafted tumor can be increased
the inoculation of murine cancer cell lines or tissues into another by serial passage. In this method, tumorigenic samples are subse-
mouse which has an identical genetic background [27,29]. Synge- quently expanded and re-implanted into the cecum for several
neic models are valuable for evaluating the interaction between generations to obtain higher tumorigenicity and metastatic abili-
tumors and their microenvironments. The graft is seldom rejected ties [19]. Such orthotopic transplantation models with highly
and they are cost-effective and convenient. However, these models metastatic potential have been demonstrated to effectively devel-
lack the genetic heterogeneity of human tumors [14,27]. In con- oped liver metastasis [38,44,45]. However, this selection process and
trast to syngeneic models, xenograft models usually refer to models the subsequent in vitro cell culture result in adaptation of tumor
developed through the inoculation of human cancer cell lines or cells to growth in a two-dimensional environment rather than in
tissues into immunodeficient mice [14,19]. These models have been the normal three-dimensional environment [27].
widely used in human cancer research, although they have limita- A major advantage of the orthotopic spontaneous liver metas-
tions such as deficiencies in immune system function and low rates tasis model is that metastatic dissemination follows a natural course
of engraftment [30–32]. In particular, patient-derived xenograft (PDX) and utilizes a mechanism like that used in humans. Thus, this model

Fig. 1. Types of mouse models for colorectal cancer with liver metastasis according to the formation method.
116 B.Y. Oh et al. / Cancer Letters 387 (2017) 114–120

Fig. 2. Transplantable models for colorectal cancer with liver metastasis. (A) Orthotopic model with serosal injection of cancer cell lines, (B) Orthotopic model with surgi-
cal implantation of cancer tissue, and (C) Experimental model with intra-splenic injection.

is suitable for predicting drug response in human tumors. This model cancer cells are directly injected into the spleen or the portal vein
represents the entire metastatic process from the formation of the (Fig. 2C). The tumor cells reach hepatic microcirculation without un-
primary site to the developing liver metastasis. Thus, it enables dergoing the steps of primary tumor growth and intravasation, and
the investigation of all steps in the metastatic cascade. It mimics are then arrested by size or specific retention factors. These ar-
the human tumor microenvironment and provides a realistic level rested cells undergo cellular extravasation and enter the liver
of heterogeneity among tumor cells. In addition, the number of pas- parenchyma or they may grow out while remaining within the vas-
sages required to isolate highly metastatic tumor cells in the culature. Some of the injected cells eventually form liver metastases
spontaneous metastasis model is less than that required for the ex- and the rest disappear without colonization in the liver. To in-
perimental metastasis model [14,19,28,46–48]. crease the efficiency of liver metastasis, the previously described
Disadvantages of this model are the low predictability and re- serial passage method is necessary in experimental models as in
producibility of spontaneous metastasis from colon to liver. It is also spontaneous models. There are several reports that highly malig-
difficult to assess the contribution of the immune system to meta- nant cells develop liver metastases when human colon cancer cells
static progression as this model due to the lack of a functional are injected into the spleens of immunocompromised mice
immune system in the xenograft. In addition, it takes a long time [12,14,19,27,49].
to develop liver metastasis, and sometimes resection of the primary Experimental metastasis models provide several advantages for
tumor is needed to reduce tumor burden and allow for the forma- metastasis research. The tumor cells are injected directly into the
tion of metastasis [18,19,28,30]. Additionally, cecal injection is systemic circulation, so it takes a short time for metastases to
technically difficult and carriers a risk of tumor cell leakage or intra- develop. Furthermore, these models are highly reproducible and con-
luminal spillage (Table 1). sistently lead to metastasis formation. Metastasis formation in these
models is more efficient compared to the spontaneous metastasis
Experimental liver metastasis models models. Use of these models enables control over the number and
types of tumor cells introduced into the circulation, which is im-
The second approach to developing liver metastasis is the in- portant in achieving the experimental endpoint. In addition, the
jection of tumor cells directly into systemic circulation, which induces metastatic organ can be targeted to specific sites [14,27,28,50,51].
liver metastasis experimentally. Experimental metastases occur in A major disadvantage of the experimental metastasis models is
different organs, depending on the site of injection and the tropism that these models cannot represent the entire metastatic process.
of the injected cell. In experimental liver metastasis models, colon They represent only the late stage of the metastasis cascade, since

Table 1
Advantages and disadvantages of models for liver metastasis according to the type of mouse model.

Model types Advantages Disadvantages

Spontaneous ● Natural course of metastatic dissemination ● Low predictability and reproducibility of metastasis
model ● Suitability for predicting drug response ● Functional immune system deficiency in xenograft
● Identification of all steps in the metastatic cascade ● Length of time required to develop liver metastasis
● Similarity to the human tumor microenvironment ● Possibility of primary tumor resection due to tumor burden
● Similarity to human disease progression ● Technical difficulties
● Maintenance of realistic heterogeneity of tumor cells ● Asynchronous development of liver metastasis
● Minimal number of passages required to obtain high metastatic potential
Experimental ● Short time required to develop liver metastasis ● Inability to represent the entire metastatic process
model ● High reproducibility and consistency of metastasis formation ● Occurrence of only the late stage of the metastasis cascade
● Targetable metastatic organs ● Necessity of serial passage to enhance tissue-specific ability
● Control over the number of cells delivered ● Artificial route of metastatic dissemination
Patient-derived ● Suitability for examining patient-specific drug response ● Low incidence of metastasis
xenograft ● Preservation of intratumoral heterogeneity of original tumor ● Length of time required to develop liver metastasis
model ● Effectiveness in evaluating the initiation and progression of metastasis ● High cost and labor-intensive
● Technical difficulties
● Immune system deficiency in xenograft
Genetically ● Identification of specific genetic mutation during carcinogenesis ● Low predictability and reproducibility of liver metastases
engineered ● Effectiveness in evaluating the early steps in tumorigenesis ● Length of time required for the development of metastasis
model ● Similarity of defined mutations to those of in human tumors ● Low incidence of metastasis
● Intact immune system of mice ● Possibility of embryonic lethality, severe developmental defects,
● Species-specific microenvironment or sterility
● Difficulty in evaluating therapeutic response
● High cost and labor-intensive
B.Y. Oh et al. / Cancer Letters 387 (2017) 114–120 117

they bypass the early stages, including primary tumor growth and proteomic spectrum of human cancers. Therefore, they provide an
intravasation of tumor cells. Tumor cells should be isolated through understanding of entire carcinogenic progress and the mecha-
serial passage to enhance their tissue-specific abilities. Metastatic nisms of specific cancer-related genes [23,61–63]. However, they
dissemination follows an artificial route, rather than its natural course usually cannot fully reproduce the genetic complexity of human
(Table 1) [14,19,27,51]. tumors.
GEMMs of colorectal cancer usually use germline or tissue-
PDX liver metastasis models wide genetic modification, which are useful in the study of hereditary
colorectal cancer [17]. However, somatic mutations develop in spo-
PDX liver metastasis models are established by implanting cancer radic colorectal cancer, which corresponds to about 80% of all
cells or tissues from patients into immunodeficient mice, in which colorectal cancer. Thus, somatically engineered mouse models are
liver metastasis can then be induced spontaneously or experimen- needed to investigate the carcinogenesis of colorectal cancer [64].
tally, as described above. Xenografts derived from cell lines are Most GEMMs for colorectal cancer have been generated by induc-
reproducible, easy to manipulate, and well characterized; however, ing mutations of adenomatous polyposis coli (APC) upon loss of
they do not exhibit tumor heterogeneity or the histopathologic and heterozygosity [65]. APC, a tumor suppressor, negatively regulates
genetic characteristics of the original tumor [52–54]. In contrast, beta-catenin concentrations and interacts with E-cadherin [66]. Al-
PDX models better reflect the characteristics and genetic diversity though APC mutations can lead to the formation of invasive colorectal
of the original tumor; thus, these models are currently the best pre- cancer, liver metastases have not been demonstrated in these models
clinical models for testing drug response, especially for patients with [65,67]. Recently, a GEMM of colorectal cancer was established that
refractory cancer. In addition, PDX models can be used for further develops liver metastases with considerable efficiency. This model
genomic and pharmacologic studies on personalized treatments was generated by Adeno-Cre injection into the colon of LSL-KRASG12V/
[53,55]. Apcflox/flox mice. Adenoviral administration of Cre into the colon
Mouse types commonly used in PDX models include nude mice, induces the loss of the APC tumor suppressor, activation of onco-
SCID mice, and NOD/SCID humanized mice [14,16]. PDX models typ- genic KRASG12V, and the generation of sporadic colorectal cancer
ically have a low engraftment rate, so co-injection of stromal followed by liver metastases. This model limits the overgrowth of
components like Matrigel or human stromal cells have been used cancer cells, therefore avoiding premature lethality, and leads to
to improve the success rate [14]. Subcutaneously transplanted PDX tumor progression and liver metastases [64].
models rarely develop liver metastasis. The most commonly used There are significant advantages to the use of GEMMs. These
PDX liver metastasis model is a patient-derived orthotopic xeno- models provide useful information about the effects of specific
graft model [45,56]. Hoffman and colleagues found that histologically genetic mutations during carcinogenesis. Compared with trans-
intact colon cancer tissues obtained surgically from patients and plantable models, GEMMs more accurately represent the natural
orthotopically implanted into the cecum of nude mice led to the course of tumor development and the interaction between tumor
development of liver metastases in PDX models after 10 passages cells and tissue microenvironments [14,16,68]. Moreover, they are
[44,45,57]. Other studies also reported that liver metastasis devel- effective for evaluating the early steps in tumorigenesis. In addi-
oped in orthotopic PDX models of colorectal cancer [55,58,59]. tion, these models have intact immune systems and species-
A major advantage of PDX metastasis models is that these models specific microenvironments [69].
can examine patient-specific therapeutic responses to drugs and A major disadvantage of genetically engineered liver metasta-
predict disease course, providing crucial information regarding per- sis models is that liver metastases seldom develop and their
sonalized treatment [16,60]. PDX models also preserve the occurrence is not predictable [19]. Even if liver metastases occur,
intratumoral heterogeneity of the original tumor at the cellular and they require a long period of time to do so [27,70]. Mutation of spe-
genetic levels compared with xenograft models derived from cancer cific genes can lead to embryonic lethality, severe developmental
cell lines [33,52,55]. In addition, PDX metastasis models provide the defects, or sterility prior to the development of metastases [24,71].
opportunity to track the initiation and progression of metastasis [60]. A potential limitation of GEMMs is that it is difficult to evaluate ther-
Despite these advantages, PDX metastasis models have several apeutic responses because these models show a low incidence of
disadvantages in metastasis research. PDX models show variable en- and take a long time to develop metastasis [72,73]. In addition, they
graftment rates and low liver metastasis rates [14,60]. The necrotic are expensive and labor-intensive (Table 1) [16,68].
area often present in patient tissues makes engraftment success even
more challenging [55]. These models are also time consuming, ex-
pensive, and technically challenging [16]. In addition, the immune Monitoring metastasis in animal models
system is impaired in PDX models, which influences cancer pro-
gression. This drawback is partially overcome by using NOD/SCID Histologic evaluation
humanized mice via injection of peripheral blood or bone marrow
cells, but even in humanized mouse models the immune system Tumor growth is measured by histologic analysis (Fig. 3). The
is not fully restored (Table 1) [16,55,60]. extent of tumor growth is determined by measuring the area of liver
tissue that has been replaced by metastatic tumor tissue [74,75].
Genetically engineered liver metastasis models This method can quantitatively determine the extent of liver me-
tastasis; however, it does have several disadvantages. A major
The other approach to developing liver metastasis is the use of disadvantage is that it can only be performed postmortem. Thus,
genetically engineered mouse models (GEMMs), which may serve it cannot be used to sequentially evaluate the process of tumor
as alternatives to human cancer xenografts. Xenografts have been growth and it is difficult to assess therapeutic response. In addi-
used to gain an understanding of human cancer progression. tion, this is a labor-intensive and time-consuming method [15,19].
However, the use of GEMMs in immunocompetent mice has been Histologic analysis is also useful in the molecular characteriza-
suggested depending on the emphasis on the importance of the tion of liver metastasis. This method can validate similarities in the
tumor microenvironment on tumor progression. GEMMs are gen- genetic or histologic features between the primary and metastatic
erated through alterations in genomic expression, such as the tumors. It is particularly suitable for investigating human tumor be-
activation of oncogenes and the inactivation of tumor suppressor havior in mice [52,76]. This supports the importance of animal
genes. They recapitulate the histologic, genomic, transcriptomic, and models as preclinical tools for metastatic biology.
118 B.Y. Oh et al. / Cancer Letters 387 (2017) 114–120

Fig. 3. Monitoring liver metastasis in animal models. (A) Histologic evaluation (macroscopic and microscopic) and (B) Imaging evaluation (MRI, PET, and bioluminescence
imaging).

Imaging evaluation Conclusion

Several imaging techniques have been developed for use in animal Mouse models for colorectal cancer with liver metastasis, such
models and, consequently, the limitations of histologic analysis can as transplantable models (spontaneous and experimental models)
be overcome (Fig. 3). In contrast to histologic analysis, imaging tech- or GEMMs, have contributed to our understanding of the meta-
niques are noninvasive and sensitive, so they can be performed static progress. The ideal liver metastasis models should permit the
several times. These techniques allow visualization of the entire predictable and reproducible establishment of liver metastasis. Fur-
process of metastasis and the response to therapy [14,19]. Imaging thermore, they should effectively represent the human tumor
is a relevant method for monitoring metastasis in animal models. microenvironment and be available to validate novel therapeutics
Conventional imaging techniques, such as magnetic resonance for cancer patients. However, there are still limitations in the gen-
imaging (MRI), computed tomography (CT), positron emission to- eration of ideal models for human liver metastasis. Fortunately, the
mography (PET), and single photon emission tomography (SPECT), development of sequence-specific genome editing techniques, such
have been widely used in monitoring metastasis formation in animal as the clustered regularly interspaced short palindromic repeats
models [77–79]. MRI and CT provide high resolution three- (CRISPR)-Cas system, provides a new means for the development
dimensional anatomical images, while PET provides high sensitivity of liver metastasis models [84–86]. Ultimately, the goal of such
and specificity for detecting metastatic lesions. MRI is particularly animal models is to design therapeutic approaches for patients with
useful in monitoring liver metastasis and therapeutic response refractory colorectal cancer. Additional research should be con-
[52,78]. ducted to develop next-generation models of human cancer, as
Currently, in vivo optical imaging techniques, such as biolumi- ultimately these efforts will promote further clinical investigation.
nescence imaging and fluorescence imaging, are being developed.
Bioluminescence imaging allows for the effective detection of tumor
Conflict of interest
cells expressing firefly luciferase, a light-emitting photoprotein. This
imaging can be performed in a sequential manner to visualize in-
None declared.
trahepatic tumor growth. In addition, it is very sensitive, fast, and
easy to manipulate [15,19]. Many studies have reported the use-
fulness of bioluminescence imaging in liver metastasis animal models Acknowledgement
[15,18,80]. Fluorescence imaging has been used to detect the growth
of tumors expressing the Aequorea victoria green fluorescent protein This study was supported by Basic Science Research Program
(GFP). This fluorescence marker is genetically-encoded and con- through the National Research Foundation of Korea (NRF)
tinuously expressed in tumor cells. Thus, it provides the ability to funded by the Ministry of Science, ICT and Future Planning
follow metastatic cells in real time [14,19,27]. Many studies have (2015R1A2A2A01003225).
monitored liver metastasis of colon cancer using this method
[81–83]. These optical imaging methods allow for monitoring of References
single metastatic cells in vivo and provide new insights into the
biology of metastasis, especially during the early stage of liver [1] M.Y. Huang, et al., DPYD, TYMS, TYMP, TK1, and TK2 genetic expressions as
metastasis. response markers in locally advanced rectal cancer patients treated with
B.Y. Oh et al. / Cancer Letters 387 (2017) 114–120 119

fluoropyrimidine-based chemoradiotherapy, Biomed Res. Int. 2013 (2013) [38] X. Fu, et al., Extensive liver metastasis from human colon cancer in nude and
931028. SCID mice after orthotopic onplantation of histologically-intact human colon
[2] Z.J. Zhang, et al., Reduced risk of colorectal cancer with metformin therapy in carcinoma tissue, Anticancer Res. 12 (5) (1992) 1395–1397.
patients with type 2 diabetes: a meta-analysis, Diabetes Care 34 (10) (2011) [39] M. Pocard, et al., Efficiency of orthotopic xenograft models for human colon
2323–2328. cancers, In Vivo 10 (5) (1996) 463–469.
[3] X. Sui, et al., Use of metformin alone is not associated with survival outcomes [40] T. Kubota, Metastatic models of human cancer xenografted in the nude mouse:
of colorectal cancer cell but AMPK activator AICAR sensitizes anticancer effect the importance of orthotopic transplantation, J. Cell. Biochem. 56 (1) (1994)
of 5-fluorouracil through AMPK activation, PLoS ONE 9 (5) (2014) e97781. 4–8.
[4] A. Jemal, et al., Global cancer statistics, CA Cancer J. Clin. 61 (2) (2011) 69– [41] M.C. Bibby, Orthotopic models of cancer for preclinical drug evaluation:
90. advantages and disadvantages, Eur. J. Cancer 40 (6) (2004) 852–857.
[5] B.Y. Oh, et al., Role of beta1-integrin in colorectal cancer: case-control study, [42] C. Khanna, et al., An orthotopic model of murine osteosarcoma with clonally
Ann. Coloproctol. 30 (2) (2014) 61–70. related variants differing in pulmonary metastatic potential, Clin. Exp. Metastasis
[6] R. Siegel, et al., Cancer statistics, 2014, CA Cancer J. Clin. 64 (1) (2014) 9–29. 18 (3) (2000) 261–271.
[7] H. Kamiyama, et al., Molecular biomarkers for the detection of metastatic [43] M.V. Cespedes, et al., Mouse models in oncogenesis and cancer therapy, Clin.
colorectal cancer cells, World J. Gastroenterol. 20 (27) (2014) 8928–8938. Transl. Oncol. 8 (5) (2006) 318–329.
[8] W. Li, et al., miRNA-99b-5p suppresses liver metastasis of colorectal cancer by [44] B. Rashidi, et al., A nude mouse model of massive liver and lymph node
down-regulating mTOR, Oncotarget 6 (27) (2015) 24448–24462. metastasis of human colon cancer, Anticancer Res. 20 (2A) (2000) 715–722.
[9] B. Lintoiu-Ursut, A. Tulin, S. Constantinoiu, Recurrence after hepatic resection [45] F.X. Sun, et al., An ultra-metastatic model of human colon cancer in nude mice,
in colorectal cancer liver metastasis – review article, J. Med. Life 8 Spec Issue Clin. Exp. Metastasis 17 (1) (1999) 41–48.
(2015) 12–14. [46] R. Munoz, et al., Highly efficacious nontoxic preclinical treatment for advanced
[10] H. Chen, et al., Silencing of plasminogen activator inhibitor-1 suppresses metastatic breast cancer using combination oral UFT-cyclophosphamide
colorectal cancer progression and liver metastasis, Surgery 158 (6) (2015) metronomic chemotherapy, Cancer Res. 66 (7) (2006) 3386–3391.
1704–1713. [47] W. Cruz-Munoz, et al., Development of a preclinical model of spontaneous
[11] H.J. Wanebo, et al., Meeting the biologic challenge of colorectal metastases, Clin. human melanoma central nervous system metastasis, Cancer Res. 68 (12) (2008)
Exp. Metastasis 29 (7) (2012) 821–839. 4500–4505.
[12] K. Ishizu, et al., Development and characterization of a model of liver metastasis [48] G. Francia, et al., Long-term progression and therapeutic response of visceral
using human colon cancer HCT-116 cells, Biol. Pharm. Bull. 30 (9) (2007) metastatic disease non-invasively monitored in mouse urine using beta-human
1779–1783. choriogonadotropin secreting tumor cell lines, Mol. Cancer Ther. 7 (10) (2008)
[13] D.M. van Marion, et al., Studying cancer metastasis: existing models, challenges 3452–3459.
and future perspectives, Crit. Rev. Oncol. Hematol. 97 (2016) 107–117. [49] I.J. Fidler, Critical factors in the biology of human cancer metastasis: twenty-
[14] M. Saxena, G. Christofori, Rebuilding cancer metastasis in the mouse, Mol Oncol eighth G.H.A. Clowes memorial award lecture, Cancer Res. 50 (19) (1990)
7 (2) (2013) 283–296. 6130–6138.
[15] N. Smakman, et al., Validation of bioluminescence imaging of colorectal liver [50] A.F. Chambers, et al., Critical steps in hematogenous metastasis: an overview,
metastases in the mouse, J. Surg. Res. 122 (2) (2004) 225–230. Surg. Oncol. Clin. N. Am. 10 (2) (2001) 243–255, vii.
[16] A. Richmond, Y. Su, Mouse xenograft models vs GEM models for human cancer [51] Y. Kang, Analysis of cancer stem cell metastasis in xenograft animal models,
therapeutics, Dis. Model. Mech. 1 (2–3) (2008) 78–82. Methods Mol. Biol. 568 (2009) 7–19.
[17] M.M. Taketo, W. Edelmann, Mouse models of colon cancer, Gastroenterology [52] Y.B. Cho, et al., Colorectal cancer patient-derived xenografted tumors maintain
136 (3) (2009) 780–798. characteristic features of the original tumors, J. Surg. Res. 187 (2) (2014)
[18] W.Y. Lee, et al., Comparison of colorectal cancer in differentially established liver 502–509.
metastasis models, Anticancer Res. 34 (7) (2014) 3321–3328. [53] E. Marangoni, et al., A new model of patient tumor-derived breast cancer
[19] M.W. Heijstek, O. Kranenburg, I.H. Borel Rinkes, Mouse models of colorectal xenografts for preclinical assays, Clin. Cancer Res. 13 (13) (2007) 3989–3998.
cancer and liver metastases, Dig. Surg. 22 (1–2) (2005) 16–25. [54] T. John, et al., The ability to form primary tumor xenografts is predictive of
[20] B. Hann, A. Balmain, Building ‘validated’ mouse models of human cancer, Curr. increased risk of disease recurrence in early-stage non-small cell lung cancer,
Opin. Cell Biol. 13 (6) (2001) 778–784. Clin. Cancer Res. 17 (1) (2011) 134–141.
[21] M.P. Rosenberg, D. Bortner, Why transgenic and knockout animal models should [55] I. Puig, et al., A personalized preclinical model to evaluate the metastatic
be used (for drug efficacy studies in cancer), Cancer Metastasis Rev. 17 (3) (1998) potential of patient-derived colon cancer initiating cells, Clin. Cancer Res. 19
295–299. (24) (2013) 6787–6801.
[22] L. He, et al., Mouse models of liver cancer: progress and recommendations, [56] Y. Hiroshima, et al., Establishment of a patient-derived orthotopic Xenograft
Oncotarget 6 (27) (2015) 23306–23322. (PDOX) model of HER-2-positive cervical cancer expressing the clinical
[23] K.K. Frese, D.A. Tuveson, Maximizing mouse cancer models, Nat. Rev. Cancer metastatic pattern, PLoS ONE 10 (2) (2015) e0117417.
7 (9) (2007) 645–658. [57] B. Rashidi, et al., An orthotopic mouse model of remetastasis of human colon
[24] K. Maddison, A.R. Clarke, New approaches for modelling cancer mechanisms cancer liver metastasis, Clin. Cancer Res. 6 (6) (2000) 2556–2561.
in the mouse, J. Pathol. 205 (2) (2005) 181–193. [58] K. Morikawa, et al., In vivo selection of highly metastatic cells from surgical
[25] M. Cekanova, K. Rathore, Animal models and therapeutic molecular targets of specimens of different primary human colon carcinomas implanted into nude
cancer: utility and limitations, Drug Des. Devel. Ther. 8 (2014) 1911–1921. mice, Cancer Res. 48 (7) (1988) 1943–1948.
[26] C.L. Morton, P.J. Houghton, Establishment of human tumor xenografts in [59] S. Julien, et al., Characterization of a large panel of patient-derived tumor
immunodeficient mice, Nat. Protoc. 2 (2) (2007) 247–250. xenografts representing the clinical heterogeneity of human colorectal cancer,
[27] C. Khanna, K. Hunter, Modeling metastasis in vivo, Carcinogenesis 26 (3) (2005) Clin. Cancer Res. 18 (19) (2012) 5314–5328.
513–523. [60] D. Siolas, G.J. Hannon, Patient-derived tumor xenografts: transforming clinical
[28] G. Francia, et al., Mouse models of advanced spontaneous metastasis for samples into mouse models, Cancer Res. 73 (17) (2013) 5315–5319.
experimental therapeutics, Nat. Rev. Cancer 11 (2) (2011) 135–141. [61] J. Carretero, et al., Integrative genomic and proteomic analyses identify targets
[29] A. Fantozzi, G. Christofori, Mouse models of breast cancer metastasis, Breast for Lkb1-deficient metastatic lung tumors, Cancer Cell 17 (6) (2010) 547–
Cancer Res. 8 (4) (2006) 212. 559.
[30] J. Jonkers, P.W. Derksen, Modeling metastatic breast cancer in mice, J. Mammary [62] E.S. Martin, et al., Development of a colon cancer GEMM-derived orthotopic
Gland Biol. Neoplasia 12 (2–3) (2007) 191–203. transplant model for drug discovery and validation, Clin. Cancer Res. 19 (11)
[31] N. Cabioglu, et al., CXCL-12/stromal cell-derived factor-1alpha transactivates (2013) 2929–2940.
HER2-neu in breast cancer cells by a novel pathway involving Src kinase [63] T. Van Dyke, T. Jacks, Cancer modeling in the modern era: progress and
activation, Cancer Res. 65 (15) (2005) 6493–6497. challenges, Cell 108 (2) (2002) 135–144.
[32] A. Orimo, et al., Stromal fibroblasts present in invasive human breast carcinomas [64] K.E. Hung, et al., Development of a mouse model for sporadic and metastatic
promote tumor growth and angiogenesis through elevated SDF-1/CXCL12 colon tumors and its use in assessing drug treatment, Proc. Natl. Acad. Sci. U.S.A.
secretion, Cell 121 (3) (2005) 335–348. 107 (4) (2010) 1565–1570.
[33] B.Y. Oh, et al., Correlation between tumor engraftment in patient-derived [65] P. Rampetsreiter, E. Casanova, R. Eferl, Genetically modified mouse models of
xenograft models and clinical outcomes in colorectal cancer patients, Oncotarget cancer invasion and metastasis, Drug Discov. Today Dis. Model 8 (2–3) (2011)
6 (18) (2015) 16059–16068. 67–74.
[34] K. Flatmark, et al., Twelve colorectal cancer cell lines exhibit highly variable [66] S.D. Markowitz, M.M. Bertagnolli, Molecular origins of cancer: molecular basis
growth and metastatic capacities in an orthotopic model in nude mice, Eur. J. of colorectal cancer, N. Engl. J. Med. 361 (25) (2009) 2449–2460.
Cancer 40 (10) (2004) 1593–1598. [67] J. Jonkers, A. Berns, Conditional mouse models of sporadic cancer, Nat. Rev.
[35] N. Guilbaud, et al., Marked antitumor activity of a new potent acronycine Cancer 2 (4) (2002) 251–265.
derivative in orthotopic models of human solid tumors, Clin. Cancer Res. 7 (8) [68] G. Dranoff, Experimental mouse tumour models: what can be learnt about
(2001) 2573–2580. human cancer immunology?, Nat. Rev. Immunol. 12 (1) (2012) 61–66.
[36] J.Y. Park, et al., Fluorescent-antibody targeting of insulin-like growth factor-1 [69] B. Firestone, The challenge of selecting the ‘right’ in vivo oncology pharmacology
receptor visualizes metastatic human colon cancer in orthotopic mouse models, model, Curr. Opin. Pharmacol. 10 (4) (2010) 391–396.
PLoS ONE 11 (1) (2016) e0146504. [70] J.E. Talmadge, et al., Murine models to evaluate novel and conventional
[37] H.N. Chen, et al., PDLIM1 stabilizes the E-cadherin/beta-catenin complex to therapeutic strategies for cancer, Am. J. Pathol. 170 (3) (2007) 793–804.
prevent epithelial-mesenchymal transition and metastatic potential of colorectal [71] R.D. Palmiter, R.L. Brinster, Germ-line transformation of mice, Annu. Rev. Genet.
cancer cells, Cancer Res. (2015). 20 (1986) 465–499.
120 B.Y. Oh et al. / Cancer Letters 387 (2017) 114–120

[72] S.G. Brodie, et al., Multiple genetic changes are associated with mammary [79] R. Weissleder, Scaling down imaging: molecular mapping of cancer in mice,
tumorigenesis in Brca1 conditional knockout mice, Oncogene 20 (51) (2001) Nat. Rev. Cancer 2 (1) (2002) 11–18.
7514–7523. [80] M.K. Nyati, et al., The potential of 5-fluorocytosine/cytosine deaminase enzyme
[73] V. Stambolic, et al., High incidence of breast and endometrial neoplasia prodrug gene therapy in an intrahepatic colon cancer model, Gene Ther. 9 (13)
resembling human Cowden syndrome in pten ± mice, Cancer Res. 60 (13) (2000) (2002) 844–849.
3605–3611. [81] T. Chishima, et al., Cancer invasion and micrometastasis visualized in live tissue
[74] E.A. te Velde, et al., Enhanced antitumour efficacy by combining conventional by green fluorescent protein expression, Cancer Res. 57 (10) (1997) 2042–
chemotherapy with angiostatin or endostatin in a liver metastasis model, Br. 2047.
J. Surg. 89 (10) (2002) 1302–1309. [82] R.M. Hoffman, Orthotopic transplant mouse models with green fluorescent
[75] Y. van Hensbergen, et al., Rapid stereology based quantitative protein-expressing cancer cells to visualize metastasis and angiogenesis, Cancer
immunohistochemistry of dendritic cells in lymph nodes: a methodological Metastasis Rev. 17 (3) (1998) 271–277.
study, Anal. Cell. Pathol. 22 (3) (2001) 143–149. [83] M. Yang, et al., Whole-body optical imaging of green fluorescent protein-
[76] L. Resor, T.J. Bowen, A. Wynshaw-Boris, Unraveling human cancer in the mouse: expressing tumors and metastases, Proc. Natl. Acad. Sci. U.S.A. 97 (3) (2000)
recent refinements to modeling and analysis, Hum. Mol. Genet. 10 (7) (2001) 1206–1211.
669–675. [84] W. Xue, et al., CRISPR-mediated direct mutation of cancer genes in the mouse
[77] P. Dubey, et al., Quantitative imaging of the T cell antitumor response by liver, Nature 514 (7522) (2014) 380–384.
positron-emission tomography, Proc. Natl. Acad. Sci. U.S.A. 100 (3) (2003) [85] F.J. Sanchez-Rivera, T. Jacks, Applications of the CRISPR-Cas9 system in cancer
1232–1237. biology, Nat. Rev. Cancer 15 (7) (2015) 387–395.
[78] J.S. Lewis, et al., Small animal imaging. Current technology and perspectives [86] H. Mou, et al., Precision cancer mouse models through genome editing with
for oncological imaging, Eur. J. Cancer 38 (16) (2002) 2173–2188. CRISPR-Cas9, Genome Med. 7 (1) (2015) 53.

You might also like