Vandamme, 2014
Vandamme, 2014
Vandamme, 2014
178]
Review Article
Use of rodents as models of human diseases
Thierry F. Vandamme
How to cite this article: Vandamme TF. Use of rodents as models of human diseases. J Pharm Bioall Sci 2014;6:2-9.
complex organisms has completely changed and enhanced the A large variety of human and murine cell lines derived from
study of all aspects of biologic processes. Transgenic mouse both, solid tumors or leukemias, covering a wide range of tumor
models in toxicology have primarily been used to screen geno‑and phenotypes, have been adapted to grow in a murine
drugs for carcinogenicity and to understand the mechanisms host and thus allow testing of a compound in the appropriate
of toxicity. These mouse models can reliably predict the tumor model [Table 1].
carcinogenic potential of compounds and significantly reduce
the risk of using these drugs in clinics to treat human diseases. The earliest xenograft models in which human tumor cells
were grown in immunosuppressed mice involved subcutaneous
Use of short‑term experiments on transgenic mice in implantation of human cell lines. Subcutaneous xenograft
combination with 2‑year chronic studies on rats could models have been popular because they are easy to establish,
increase the overall accuracy of detecting carcinogens and easy to manage and lend themselves to ready quantitation of
non‑carcinogens. Testing new drugs using different species also the tumor burden. More recently, orthotopic xenograft models,
reduces false results. Additional advantages of using transgenic in which the tumor cells are implanted in the tumor site of
assays include reduced duration of studies, conservative use of origin, have been used with greater frequency in animal studies
animals and lower cost relative to a traditional 2‑year rodent of cancers. Orthotopic xenograft models are advantageous for
chronic toxicity study (http://www.ruro.com/blog/3752, viewed their ability to mimic local tumor growth and recapitulate the
27th January 2013). pathways of metastasis seen in human cancers. In addition,
recent innovations in cell labeling techniques and small‑animal
Xenografts imaging have enabled investigators to monitor the metastatic
process and quantitate the growth and spread of orthotopically
Xenografts (xenos‑from the Greek meaning “foreign”), is a graft implanted tumors.[36]
obtained from a member of one species and transplanted to a
member of another species, genus, or family. The transplantation Progress in oncology drug development has been hampered by
can consist of living cells, tissues or organs from one species to a lack of preclinical models that reliably predict clinical activity
another. Xenografts are used to answer key questions in the of novel compounds in cancer patients. In an effort to address
field of cancer research when it is necessary to rely upon the these shortcomings, there has been a recent increase in the
use of animal model systems that closely resemble tumor use of patient‑derived tumor xenografts (PDTX), engrafted
progression in the human patient. Human xenografts growing into immune‑compromised rodents such as athymic nude or
in immunodeficient mice are a well‑established and useful non‑obese diabetic/SCID mice, for use as preclinical models.
model for studying human tumor biology in a system that better
mimics the primary tumor as compared to cells grown in vitro.
Often times, xenograft studies use highly passaged cell lines that Table 1: Subcutaneous in vivo tumor cell lines models
have been genetically modified and artificially cultured, leading Tumor origin Tumor cell lines Species References
to a clonal selection that may not be observed in patients. Ideal Blood/leukemia MV4‑11 Human [6]
primary tumor tissue xenografts result from patient‑derived MOLM‑13 Human [7]
passes removed from patient). Furthermore, these cell lines Bone SJSA‑1 Human [9]
tumor tissue at low passage is believed to conserve original Cervix C33A Human [13]
biomolecular signature, malignant phenotypes and genotypes, Colon HCT‑116 Human [15]
prevalent hypothesis, primary tumor xenografts are believed to CT26 Mouse [18]
offer relevant predictive insights into clinical outcomes when LS 174T Human [19]
most commonly performed. It is also the fastest and lowest cost Lung H460 Human [25]
model. In these subcutaneous models, the tumor is grown under A549 Human [26]
are generally injected subcutaneously into immunodeficient Prostate PC‑3 Human [31]
mice, which are then treated with the compound of interest LNCaP Human [32]
for 2‑6 weeks during which time subcutaneous tumors develop. Skin A431 Human [33]
The study is “positive” if the compound of interest reduces the A375 Human [34]
Numerous tumor‑specific PDTX models have been established of efficient and targeted treatments (e.g., leukemia,
and importantly, they are biologically stable when passaged in hypertension and obesity).
mice in terms of global gene‑expression patterns, mutational
status, metastatic potential, drug responsiveness and tumor The major applications for genetically modified animals in
architecture. These characteristics might provide significant agriculture are
improvements over standard cell‑line xenograft models.[5] • To generate animal with desired breeding traits (e.g., lower
phosphorous in the dung).
Genetically Engineered Animal Models • To induce resistance against disease (e.g., fish farming) (Basel
Declaration, 2010).
Because the genetics and histology of xenografts do not
recapitulate the genetics and histology of human tumors, One of the goals in using transgenic animals is that it allows for
genetically engineered animal models were developed. a reduction in the use of larger animals. The use of GEMs also
provides an opportunity to improve our understanding of the
The use of genetically modified mice for carcinogenicity mechanisms of action for potential therapeutic compounds. At
evaluation began more than 20 years ago, when researchers present, the European Commission consider that mice appear
found that different strains of genetically engineered mice to be the most common genetically engineered animal models
demonstrated that cancer incidence is increased and tumor to study new drug compounds for different diseases for the
latency is decreased in mice whose germ line, the Ha‑ras following reasons[42]
oncogene, has been inserted. Previously, the selection of mouse • The mouse is the most common model organism for
oncology models was based simply on availability of a mouse preclinical studies even though it has not proven particularly
strain and a known compatible tumor. This frequently resulted reliable at predicting the outcome of studies in humans.
in the use of tumor models that while long on history were short • Mice have many advantages over other model organisms:
on homology and quality control. For these reasons, preclinical Their genome is similar to the human genome (99%), a good
efficacy testing for anti‑tumor therapies should progress through genetic/molecular toolbox is available and the animal’s small
a series of models of increasing sophistication that includes size facilitates large scale/high throughput studies making it
incorporation of genetically engineered animals and orthotopic a cost‑efficient model. Therefore, it’s potential for making
and combination therapy models. medical research and in particular drug development more
efficient could be increased by solving a range of identified
Genetically modified animals are organisms in which specific bottlenecks.
genes have been altered (added or ablated) to create models for • Mouse models have been successfully used to validate
human and animal diseases. A transgenic animal is defined as drug targets and to determine efficacious and safe dosage
an animal that carries one or more foreign genes, deliberately schemes for combination treatments in humans. These
introduced through insertion into the animal’s genome. The cases have one factor in common: They do not aim to fully
foreign gene is constructed using recombinant deoxyribonucleic model a disease or disease mechanisms, but rather set out
acid (DNA) technologies. The introduction of a gene can also to obtain specific functional information.
generate therapeutic medicinal products. Standard genetically • Genetically modified mice need to be validated, reproducible,
modified animals include laboratory flies, fish, worms, rodents robust and cost‑effective to be considered optimal by the
and (for agricultural or production purposes) pigs, sheep and pharmaceutical industry.
cows. In no case is genetic modification of man implied.[37,38] • Transgenic humanized mice could provide good preclinical
screening and safety testing models for use in lead
In a modern GEM models, oncogenes are activated and/or identification and optimization.
tumor‑suppressor genes (TSGs) are inactivated somatically, • The large‑scale phenotyping of genetically engineered mice
generally through temporally controlled and tissue‑specific can provide valuable information on gene function which
expression of CRE recombinase (tyrosine recombinase is relevant for human health and disease.
enzyme derived from the P1 Bacteriophage). Animals then • The use of mice in clinical studies has proven effective
develop tumors in the tissue of interest (in this case the lung). in a number of cases. Dosage regimes of new treatments
Tumor‑bearing genetically engineered mice are then treated with or treatment combinations can rapidly be optimized by
the compound of interest and serially assessed for response.[39] co‑clinical trials with mice, allowing fast application in
humans with greater patient safety. This provides increasing
The benefits of engineered animal models are numerous. opportunities in particular for the rapid development of
Examples include[40,41] treatments for very rare diseases where low patient numbers
• Development and testing of safe and effective products for otherwise hamper the creation of clinical trials.
human application (e.g., human antibodies).
• The production of recombinant products (anti‑coagulant; One of the key bottlenecks that needs to be solved concerns
therapeutic antibodies). the aggressive patenting strategy (including the patenting of
• A means to study disease mechanisms in a complex mouse genes and broad based methods patents) and overly
organism (diabetes). restrictive licensing terms in material transfer agreements, which
• Understanding the mechanistic causes and pathways hamper the construction, sharing, use and proper exploitation of
underlying human disease, to permit the development mouse models. Representatives from the European Medicines
Agency and the Food and Drug Administration confirmed that and bioluminescence imaging. These new technologies will
data stemming from mouse models will be taken into account hopefully support the use of GEMs in preclinical trials and help
wherever the relevance has been clearly proven within the given determine if trials in GEMs are more predicative than xenografts
context. of human responses.[43]
The efficient development and use of these mouse models The selection of murine cancer models is often based simply on
necessitates the collaboration of experts within academia, the availability of a mouse strain and on a known compatible
clinicians and industry. In this regard, there is a need for tumor. Frequently this information results in use of tumor
specialist training programs that provide venue whereby insights models long on history, but short on homology and quality
can be shared from mouse pathologists, human pathologists control. Other factors need to be considered, including
and clinicians in order to better define the opportunities and genetics, sex, immunological status, method and site of tumor
limitations associated with use of these mouse models. implantation, technical competence and biological activity
of the tumor, protocol sequence and timing and selection of
The Use of Mouse Models to Support Therapeutic endpoints. Each of these variables can influence the data derived
Drug Development from use of these tumor models.
Animal models are being increasingly used in the study of disease It is frequently observed that murine tumor models error
progression and for safety assessments of new compounds. They towards false positive therapeutic results, curing the cancer in
are a powerful tool for developing a more detailed understanding mice but not in humans. This perceived inadequacy of classical
of the role specific genes play in biological pathways. At the transplantation models for the development of anti‑tumor
present time, the ability to introduce new genetic information therapies is currently a pivotal factor in driving the development
into the germ line of complex organisms has completely changed and use of GEMs. The underlying limitations of tumor
and enhanced the study of all aspects of biologic processes. models also serve to reinforce the need for careful attention to
design (applying correct models to the question), conduct (using
Cancer multiple models) and interpretation (recognizing limitations
and applying stringent criteria to outcomes) of efficacy
Mouse models of human cancer are valuable tools for cancer studies for tumor modulation. Thus, while animal models
research. Although xenografts and GEMs possess limitations can provide a form of “high throughput” in the selection of
as well as advantages, each system plays a significant role in potential drug gable candidates, the use of these results to
preclinical testing. predict human clinical outcomes is premature. Therefore,
new strategies, techniques and continued improvements in
Tumor xenografts are easy to use, relatively inexpensive stringency and consistency of criteria used for evaluating
and reproducible. The main drawback of xenografts is that outcomes are necessary to ensure that tumor models in mice
the genetics and histology of the tumors are frequently not remain a useful tool for development of anticancer agents and
representative of the respective human tumor and thus far, devices.[44] Moreover, transitions from in vitro to preclinical and
these models have not been as predictive of therapeutic success then to clinical testing for tumor modulation remain difficult
as one would like.[43] The use of xenografts to design novel with a low rate of clinical entry for most therapeutic classes.
specific anti‑tumor therapies for enhanced and targeted drug Increased understanding of mechanisms of neoplasia through
delivery such as anti‑angiogenesis (inhibitors and enhanced macromolecular biology, genomics and bioinformatics is helping
permeation), immunotherapy (vaccines, monoclonal antibodies, to address treatment bottlenecks. These bottlenecks include a
toxin conjugates, prodrug activators, cytokine antagonists), lack of specificity, low efficacy, toxicity and drug resistance and
small molecules (inhibitors of growth, matrix and adhesion), the need to identify critical targets for clinical exploitation.
apoptosis (enhancers, inducers, proteasome inhibitors, reverse
DNA methylation), anti‑sense and gene therapy (TSGs) and The numerous studies evaluating new antitumor drugs use
cell cycle alterations (inhibitors) are not sufficient to support engineered animal models [Table 2].
their use as the sole drug screen in an anti‑tumor evaluation.
Therefore, to support the development of cancer therapeutics, Other applications
there needs to be a series of models used, including GEMs and
orthotopic and combination therapy models. Intestinal inflammation
In contrast to studies involving xenografts, GEMs are Rennick and Fort used interleukin (IL)‑10‑/‑mice, which
histologically and genetically accurate models of human spontaneously develop this pathological condition that
cancer but have disadvantages of heterogeneity with regard is characterized by discontinuous transmural lesions
to frequency, latency and growth. These disadvantages are affecting the small and large intestine. This inflammatory
reminiscent of the variable behavior of actual human tumors. response also includes the dysregulated production of
Recently, these shortcomings have been partly overcome with proinflammatory cytokines.[52] The uncontrolled generation of
the development of anatomic and molecular in vivo imaging interferon‑gamma‑producing CD41 T‑cells (Th1 type) has been
techniques such as magnetic resonance imaging (MRI) shown to play a causal role in the development of enterocolitis
HER2‑positive breast carcinomas Vitamin E analogs Breast tumor mouse model [46]
MPEG: Monomethoxy poly(ethylene glycol), PCL: Poly(ε‑caprolactone), EGFP: Enhanced green fluorescent protein, HER2: Human epidermal growth
factor receptor 2, TRAMP: Transgenic adenocarcinoma mouse prostate, VEGFR2: Vascular endothelial growth factor receptor 2, HCC: Hepatocellular
carcinoma
affecting these mutants. the study of IL‑102/2 mice have been unrelated to transamidase catalytic activity. Iismaa et al.[54] in
applied to an evaluation of the role of enteric organisms in their study reviewed recent insights into the physiology and
triggering intestinal disease, the mediators responsible for pathophysiology of TG family members that have come from
initiating and maintaining intestinal disease, the role IL‑10 studies of GEM models and/or inherited disorders. The review
plays in the generation and/or function of regulatory cells and focused on FXIII‑A, TG1, TG2, TG5 and protein 4.2, as mice
the results of IL‑10 therapy in experimental animal models deficient in TG3, TG4, TG6, or TG7 and they underlined the
of inflammatory bowel disease (IBD) and human patients necessity to use these engineered animals to study these proteins
with IBD. linked to human disease.
Although rodent models do not always perfectly reproduce the Recently, Jean‑Pierre Changeux underlined the interest in using
human pathology of non‑alcoholic fatty liver disease (NAFLD), transgenic animals to study nicotine addiction and nicotinic
Postic and Girard concluded that the use of transgenic, receptors. Indeed, in an interesting review, he mentions that
knockout and knockdown mouse models have helped over recent studies in mice involving deletion and replacement of
the past years to better the understanding of the molecular nicotinic acetylcholine receptor subunits have begun to identify
determinants of NAFLD. Key enzymes of fatty acid synthesis, the molecular mechanisms underlying nicotine addiction and
such as acetyl‑CoA carboxylase, elongation of long‑chain fatty might offer new therapeutic strategies to treat this addiction.[55]
acids family member 6, stearoyl-Coenzyme A desaturase 1
(SCD1), glycerol‑3‑phosphate acyltransferase, or diacylglycerol Arachidonic acid metabolism and central nervous system
acyltransferase, have been shown, when knocked down, to pathology
reverse many of the metabolic defects associated with hepatic
steatosis and/or insulin resistance, indicating that decreased Bosetti used genetic mouse models to investigate the
TG synthesis in liver is a potential and interesting target for physiological and pathological roles of Arachidonic acid (AA)
the treatment of NAFLD.[53] Therefore, a better understanding release and metabolism in brain.[56] She concluded that although
of the function and/or regulation of the transcription factors data obtained in genetically altered mouse models should be
that control the activity of the enzymes modulating fatty interpreted with care because of possible compensatory changes,
acid synthesis in liver, namely carbohydrate response element knockout and transgenic mice provide a useful approach to
binding protein, sterol regulatory element binding protein‑1c identify the roles of specific enzymes of the AA cascade in
and liver X receptors, may in the future help the development physiological and pathological models. Therefore, these genetic
of potential therapeutic approaches for this disease animal models combined with the use of conditional knockouts,
in vivo ribonucleic acid interference and specific pharmacological
Patho‑physiology of transglutaminase inhibitors, may help to develop novel therapeutic strategies for
diseases involving altered brain AA metabolism.
The human TG family consists of a structural protein, protein
4.2, that lacks catalytic activity and eight zymogens/enzymes, AA is freed from a phospholipid molecule by the enzyme
designated factor XIII‑A (FXIII‑A) and TG1‑7, that catalyze phospholipase A2 (PLA2), which cleaves off the fatty acid,
three types of posttranslational modification reactions: but can also be generated from diacylglycerol by diacylglycerol
Transamidation, esterification and hydrolysis. These reactions lipase. AA is generated for signaling purposes appears to
are essential for biological processes such as blood coagulation, be derived by the action of a phosphatidylcholine‑specific
skin barrier formation and extracellular matrix assembly. cytosolic PLA2 (85 kDa), whereas inflammatory AA is
However, they can also contribute to the pathophysiology generated by the action of a low molecular‑weight secretory
of various inflammatory, autoimmune and degenerative PLA2 (14‑18 kDa). AA is a precursor in the production of
conditions. Some members of the TG family, for example, eicosanoids: (i) The enzymes cyclooxygenase and peroxidase
TG2, can participate in biological processes through actions lead to prostaglandin H2, which in turn is used to produce
the prostaglandins, prostacyclin and thromboxanes; (ii) the therapeutics. Indeed, deficiencies in the standard preclinical
enzyme 5‑lipoxygenase leads to 5‑HPETE, which in turn methods for evaluating potential bioactive drugs, such as
is used to produce the leukotrienes; (iii) AA is also used in xenograft mouse models, have been highlighted as a key
the biosynthesis of anandamide; (iiii) some AA is converted obstacle in the translation of the major advances in basic
into hydroxyeicosatetraenoic acids and epoxyeicosatrienoic therapeutic research into meaningful clinical benefits. At
acids by epoxygenase. The production of these derivatives present, uses of xenograft mouse models for different drug
and their action in the body are collectively known as the developments appear to be limited. In contrast, opportunities
“AA cascade”. for applying GEM models more faithfully mimic biological
evolution of different human diseases. Greater use of such
Ovarian endometrioid adenocarcinoma GEMs for preclinical studies in target validation, assessment
of disease response, investigation of pharmacodynamic
Using the combination of an OEA GEM and GEM of OEA markers of drug action, the modeling of drug resistance and
of molecular‑imaging technologies, Wang et al. studied the for understanding drug toxicity can each potentially improve
activation of the AKT serine/threonine kinase in response the likelihood of successful drug development. Incorporating a
to long‑term cisplatin therapy.[57] The authors showed that 6‑month transgenic mouse model into safety testing strategies
the treatment of cells in culture and tumor‑bearing animals for new drugs/chemicals makes valid scientific, ethical and
with cisplatin resulted in activation of AKT, a key mediator of sound business sense, since these assays are shorter in duration,
cell survival. On the basis of these results, they investigated use fewer animals and the cost is well below the traditional
the therapeutic use of AKT inhibition in combination with 2‑year mouse bioassay.
cisplatin, which resulted in enhanced and prolonged induction
of apoptosis and in significantly improved tumor control when References
compared with either agent alone. They concluded that their
results provide an impetus for clinical trials using combination 1. Workman P, Aboagye EO, Balkwill F, Balmain A, Bruder G, Chaplin DJ,
et al. Guidelines for the welfare and use of animals in cancer research.
therapy. To facilitate these trials, they also showed the use of Br J Cancer 2010;102:1555‑77.
diffusion‑weighted MRI as an imaging biomarker for evaluation 2. Angstadt AY, Thayanithy V, Subramanian S, Modiano JF, Breen M.
of therapeutic efficacy in OEA. A genome‑wide approach to comparative oncology: High‑resolution
oligonucleotide aCGH of canine and human osteosarcoma pinpoints
shared microaberrations. Cancer Genet 2012;205:572‑87.
Pancreatic cancer 3. Hansen K, Khanna C. Spontaneous and genetically engineered animal
models; use in preclinical cancer drug development. Eur J Cancer
The lack of early detection and the use of ineffective 2004;40:858‑80.
4. Cree IA, Glaysher S, Harvey AL. Efficacy of anti‑cancer agents in cell
interventions are major factors contributing to the poor
lines versus human primary tumour tissue. Curr Opin Pharmacol
prognosis and dismal survival rates of pancreatic cancer 2010;10:375‑9.
patients for more than 60 years. Detection of pancreatic 5. Tentler JJ, Tan AC, Weekes CD, Jimeno A, Leong S, Pitts TM, et al.
cancer at an early stage might permit life‑saving intervention. Patient‑derived tumour xenografts as models for oncology drug
development. Nat Rev Clin Oncol 2012;9:338‑50.
Clinical and preclinical diagnosis and evaluation of pancreatic 6. Batra V, Maris JM, Kang MH, Reynolds CP, Houghton PJ, Alexander D,
cancers involve several imaging technologies MRI, positron et al. Initial testing (stage 1) of SGI‑1776, a PIM1 kinase inhibitor,
emission tomography, computed tomography, ultrasound, by the pediatric preclinical testing program. Pediatr Blood Cancer
bioluminescent imaging and single photon emission computed 2012;59:749‑52.
7. McCormack E, Haaland I, Venås G, Forthun RB, Huseby S, Gausdal G,
tomography. The advent of genetically engineered animal et al. Synergistic induction of p53 mediated apoptosis by valproic acid
models that recapitulate the cellular and molecular pathology and nutlin‑3 in acute myeloid leukemia. Leukemia 2012;26:910‑7.
of human pancreatic intraepithelial neoplasia and pancreatic 8. Yun SH, Park ES, Shin SW, Na YW, Han JY, Jeong JS, et al. Stichoposide
C induces apoptosis through the generation of ceramide in leukemia
ductal adenocarcinoma (PDAC) has not yet yielded translational
and colorectal cancer cells and shows in vivo antitumor activity. Clin
implications. Although the use of tumor xenografts to predict Cancer Res 2012;18:5934‑48.
drug efficacy in patients has been disappointing, use of novel 9. Beltran PJ, Chung YA, Moody G, Mitchell P, Cajulis E, Vonderfecht S,
transgenic mice models should permit improved early detection et al. Efficacy of ganitumab (AMG 479), alone and in combination with
rapamycin, in Ewing’s and osteogenic sarcoma models. J Pharmacol
and development of drug regimens through integration of Exp Ther 2011;337:644‑54.
appropriate imaging modalities. In a very descriptive review, 10. Yu Y, Gao S, Li Q, Wang C, Lai X, Chen X, et al. The FGF2‑binding
Mohammed et al. [58] consider issues that are unique to peptide P7 inhibits melanoma growth in vitro and in vivo. J Cancer
working with transgenic mouse models, such as the biology of Res Clin Oncol 2012;138:1321‑8.
11. Chughtai K, Jiang L, Greenwood TR, Glunde K, Heeren RM. Mass
GEM models, stage‑tumor‑specific detection using imaging spectrometry images acylcarnitines, phosphatidylcholines, and
technologies, use of monoclonal antibodies, nanoparticles sphingomyelin in MDA‑MB‑231 breast tumor models. J Lipid Res
and biomarkers and development of chemopreventive and 2013;54:333‑44.
12. Martin LA, Pancholi S, Farmer I, Guest S, Ribas R, Weigel MT, et al.
chemotherapeutic drugs for PDAC.
Effectiveness and molecular interactions of the clinically active
mTORC1 inhibitor everolimus in combination with tamoxifen or
Conclusion letrozole in vitro and in vivo. Breast Cancer Res 2012;14:R132.
13. Chang JT, Kuo TF, Chen YJ, Chiu CC, Lu YC, Li HF, et al. Highly
potent and specific siRNAs against E6 or E7 genes of HPV16‑ or
The absence of effective in vivo systems that accurately HPV18‑infected cervical cancers. Cancer Gene Ther 2010;17:827‑36.
predict clinical efficacy has hindered drug development in 14. Chen Y, Ma J, Wang F, Hu J, Cui A, Wei C, et al . Amygdalin
induces apoptosis in human cervical cancer cell line HeLa cells. 33. Cheng KW, Mattheolabakis G, Wong CC, Ouyang N, Huang L,
Immunopharmacol Immunotoxicol 2013;35:43‑51. Constantinides PP, et al. Topical phospho‑sulindac (OXT‑328) is
15. Tu Z, Ma Y, Tian J, Li H, Akers W, Achilefu S, et al. Estrogen receptor effective in the treatment of non‑melanoma skin cancer. Int J Oncol
β potentiates the antiproliferative effect of raloxifene and affects the 2012;41:1199‑203.
cell migration and invasion in HCT‑116 colon cancer cells. J Cancer 34. Kamran MZ, Gude RP. Preclinical evaluation of the antimetastatic
Res Clin Oncol 2012;138:1091‑103. efficacy of Pentoxifylline on A375 human melanoma cell line. Biomed
16. Lin J, Yu Y, Shigdar S, Fang DZ, Du JR, Wei MQ, et al. Enhanced antitumor Pharmacother 2012;66:617‑26.
efficacy and reduced systemic toxicity of sulfatide‑containing 35. Yu K , Chen Z , Pan X , Yang Y, Tian S, Zhang J, et al .
nanoliposomal doxorubicin in a xenograft model of colorectal cancer. Tetramethylpyrazine‑mediated suppression of C6 gliomas involves
PLoS One 2012;7:e49277. inhibition of chemokine receptor CXCR4 expression. Oncol Rep
17. Yin P, Wang Y, Qiu Y, Hou L, Liu X, Qin J, et al. Bufalin‑loaded 2012;28:955‑60.
mPEG‑PLGA‑PLL‑cRGD nanoparticles: Preparation, cellular uptake, 36. Sano D, Myers JN. Xenograft models of head and neck cancers. Head
tissue distribution, and anticancer activity. Int J Nanomedicine Neck Oncol 2009;1:32.
2012;7:3961‑9. 37. Blandini F, Armentero MT. Animal models of Parkinson’s disease.
18. Zhang H, Zhang S, He H, Zhao W, Chen J, Shao RG. GAP161 targets FEBS J 2012;279:1156‑66.
and downregulates G3BP to suppress cell growth and potentiate 38. Forabosco F, Löhmus M, Rydhmer L, Sundström LF. Genetically
cisplaitin‑mediated cytotoxicity to colon carcinoma HCT116 cells. modified farm animals and fish in agriculture: A review. Livestock
Cancer Sci 2012;103:1848‑56. Sci 2013;153:1‑9.
19. Hsu CW, Chang YJ, Chang CH, Chen LC, Lan KL, Ting G, 39. Sharpless NE, Depinho RA. The mighty mouse: Genetically
et al . Comparative therapeutic efficacy of rhenium‑188 engineered mouse models in cancer drug development. Nat Rev
radiolabeled‑liposome and 5‑fluorouracil in LS‑174T human colon Drug Discov 2006;5:741‑54.
carcinoma solid tumor xenografts. Cancer Biother Radiopharm 40. Jones TS, Holland EC. Animal models for glioma drug discovery.
2012;27:481‑9. Expert Opin Drug Discov 2011;6:1271‑83.
20. Burden RE, Gormley JA, Kuehn D, Ward C, Kwok HF, Gazdoiu M, 41. Flisikowska T, Kind A, Schnieke A. The new pig on the block:
et al . Inhibition of Cathepsin S by Fsn0503 enhances the Modelling cancer in pigs. Transgenic Res 2013;22:673‑80.
efficacy of chemotherapy in colorectal carcinomas. Biochimie 42. European Commission. Of mice and men – Are mice relevant
2012;94:487‑93. models for human disease? Outcomes of the European Commission
21. James J, Ruggeri B, Armstrong RC, Rowbottom MW, Jones‑Bolin S, workshop “Are mice relevant models for human disease?” held in
Gunawardane RN, et al . CEP‑32496: A novel orally active London, UK, on May 2010.
BRAF (V600E) inhibitor with selective cellular and in vivo antitumor 43. Becher OJ, Holland EC. Genetically engineered models have
activity. Mol Cancer Ther 2012;11:930‑41. advantages over xenografts for preclinical studies. Cancer Res
22. Jagoda EM, Lang L, Bhadrasetty V, Histed S, Williams M, 2006;66:3355‑8, 3358.
Kramer‑Marek G, et al. Immuno‑PET of the hepatocyte growth factor 44. Schuh JC. Trials, tribulations, and trends in tumor modeling in mice.
receptor Met using the 1‑armed antibody onartuzumab. J Nucl Med Toxicol Pathol 2004;32 Suppl 1:53‑66.
2012;53:1592‑600. 45. Gong C, Deng S, Wu Q, Xiang M, Wei X, Li L, et al. Improving
23. Wada Y, Yoshida K, Suzuki T, Mizuiri H, Konishi K, Ukon K, et al. antiangiogenesis and anti‑tumor activity of curcumin by biodegradable
Synergistic effects of docetaxel and S‑1 by modulating the expression polymeric micelles. Biomaterials 2013;34:1413‑32.
of metabolic enzymes of 5‑fluorouracil in human gastric cancer cell 46. Dong LF, Grant G, Massa H, Zobalova R, Akporiaye E, Neuzil J.
lines. Int J Cancer 2006;119:783‑91. α‑Tocopheryloxyacetic acid is superior to α‑tocopheryl succinate in
24. Ellis L, Shah P, Hammers H, Lehet K, Sotomayor P, Azabdaftari G, et al. suppressing HER2‑high breast carcinomas due to its higher stability.
Vascular disruption in combination with mTOR inhibition in renal cell Int J Cancer 2012;131:1052‑8.
carcinoma. Mol Cancer Ther 2012;11:383‑92. 47. Mueller M, Reichardt W, Koerner J, Groettrup M. Coencapsulation
25. Wang Y, Su HH, Yang Y, Hu Y, Zhang L, Blancafort P, et al. Systemic of tumor lysate and CpG‑ODN in PLGA‑microspheres enables
delivery of modified mRNA encoding herpes simplex virus 1 successful immunotherapy of prostate carcinoma in TRAMP mice.
thymidine kinase for targeted cancer gene therapy. Mol Ther J Control Release 2012;162:159‑66.
2013;21:358‑67. 48. Zhong ZF, Hoi PM, Wu GS, Xu ZT, Tan W, Chen XP, et al. Anti‑angiogenic
26. Xie H, Lee MH, Zhu F, Reddy K, Peng C, Li Y, et al. Identification of effect of furanodiene on HUVECs in vitro and on zebrafish in vivo.
an Aurora kinase inhibitor specific for the Aurora B isoform. Cancer J Ethnopharmacol 2012;141:721‑7.
Res 2013;73:716‑24. 49. van den Engel NK, Rüttinger D, Rusan M, Kammerer R, Zimmermann W,
27. Gerber DE, Gupta P, Dellinger MT, Toombs JE, Peyton M, Hatz RA, et al. Combination immunotherapy and active‑specific tumor
Duignan I, et al. Stromal platelet‑derived growth factor receptor cell vaccination augments anti‑cancer immunity in a mouse model
α (PDGFRα) provides a therapeutic target independent of tumor of gastric cancer. J Transl Med 2011;9:140.
cell PDGFRα expression in lung cancer xenografts. Mol Cancer Ther 50. Hassan S, Buchanan M, Jahan K, Aguilar‑Mahecha A, Gaboury L,
2012;11:2473‑82. Muller WJ, et al. CXCR4 peptide antagonist inhibits primary breast
28. Heyerdahl H, Abbas N, Brevik EM, Mollatt C, Dahle J. Fractionated tumor growth, metastasis and enhances the efficacy of anti‑VEGF
therapy of HER2‑expressing breast and ovarian cancer xenografts in treatment or docetaxel in a transgenic mouse model. Int J Cancer
mice with targeted alpha emitting 227Th‑DOTA‑p‑benzyl‑trastuzumab. 2011;129:225‑32.
PLoS One 2012;7:e42345. 51. Bergé M, Bonnin P, Sulpice E, Vilar J, Allanic D, Silvestre JS,
29. Wang H, Galbán S, Wu R, Bowman BM, Witte A, Vetter K, et al. et al . Small interfering RNAs induce target‑independent
Molecular imaging reveals a role for AKT in resistance to cisplatin inhibition of tumor growth and vasculature remodeling in
for ovarian endometrioid adenocarcinoma. Clin Cancer Res a mouse model of hepatocellular carcinoma. Am J Pathol
2013;19:158‑69. 2010;177:3192‑201.
30. Rosa R, Melisi D, Damiano V, Bianco R, Garofalo S, Gelardi T, et al. 52. Rennick DM, Fort MM. Lessons from genetically engineered animal
Toll‑like receptor 9 agonist IMO cooperates with cetuximab in models. XII. IL‑10‑deficient (IL‑10(‑/‑) mice and intestinal inflammation.
K‑ras mutant colorectal and pancreatic cancers. Clin Cancer Res Am J Physiol Gastrointest Liver Physiol 2000;278:G829‑33.
2011;17:6531‑41. 53. Postic C, Girard J. Contribution of de novo fatty acid synthesis to
31. Choi ES, Jung JY, Lee JS, Park JH, Cho NP, Cho SD. Myeloid cell hepatic steatosis and insulin resistance: Lessons from genetically
leukemia‑1 is a key molecular target for mithramycin A‑induced engineered mice. J Clin Invest 2008;118:829‑38.
apoptosis in androgen‑independent prostate cancer cells and a tumor 54. Iismaa SE, Mearns BM, Lorand L, Graham RM. Transglutaminases
xenograft animal model. Cancer Lett 2013;328:65‑72. and disease: Lessons from genetically engineered mouse models
32. Authier S, Tremblay S, Dumulon V, Dubuc C, Ouellet R, Lecomte R, and inherited disorders. Physiol Rev 2009;89:991‑1023.
et al. [11C] acetoacetate utilization by breast and prostate tumors: 55. Changeux JP. Nicotine addiction and nicotinic receptors: Lessons
A PET and biodistribution study in mice. Mol Imaging Biol from genetically modified mice. Nat Rev Neurosci 2010;11:389‑401.
2008;10:217‑23. 56. Bosetti F. Arachidonic acid metabolism in brain physiology and
pathology: Lessons from genetically altered mouse models. of genetically engineered mouse models and advanced imaging
J Neurochem 2007;102:577‑86. technologies. Curr Med Chem 2012;19:3701‑13.
57. Wang X, Deng Y, Mao Z, Ma X, Fan X, Cui L, et al. CCN1 promotes
tumorigenicity through Rac1/Akt/NF‑κB signaling pathway in
pancreatic cancer. Tumour Biol 2012;33:1745‑58.
58. Mohammed A, Janakiram NB, Lightfoot S, Gali H, Vibhudutta A, Source of Support: Nil, Conflict of Interest: None declared.
Rao CV. Early detection and prevention of pancreatic cancer: Use