You are on page 1of 5

Oral Oncology 73 (2017) 16–20

Contents lists available at ScienceDirect

Oral Oncology
journal homepage: www.elsevier.com/locate/oraloncology

Review

Current mouse models of oral squamous cell carcinoma: Genetic and


chemically induced models
Kazuhisa Ishida a,b, Hiroyuki Tomita a,⇑, Takayuki Nakashima a,b, Akihiro Hirata c, Takauji Tanaka d,
Toshiyuki Shibata b, Akira Hara a
a
Department of Tumor Pathology, Gifu University Graduate School of Medicine, 1-1 Yanagido, Gifu 501-1194, Japan
b
Department of Oral Maxillofacial Surgery, Gifu University Graduate School of Medicine, 1-1 Yanagido, Gifu 501-1194, Japan
c
Division of Animal Experiment, Life Science Research Center, Gifu University, 1-1 Yanagido, Gifu 501-1194, Japan
d
Department of Diagnostic Pathology (DDP) and Research Center of Diagnostic Pathology (RC-DiP), Gifu Municipal Hospital, 7-1 Kashima-cho, Gifu City, Gifu 500-8513, Japan

a r t i c l e i n f o a b s t r a c t

Article history: Oral squamous cell carcinoma (OSCC) patients have a low 5-year survival rate and poor prognosis. To
Received 2 December 2016 improve survival and prognosis, the causes and processes involved in lesion development should be eval-
Received in revised form 11 July 2017 uated. For this purpose, the use of OSCC mouse models, such as chemically induced mouse models, genet-
Accepted 28 July 2017
ically modified mouse models, and transplanted (xenograft) models, is crucial. These OSCC models
exhibit both advantages and disadvantages when studying OSCC development and progression. Until a
model resembling human OSCC is developed, both the advantages and disadvantages of each model
Keywords:
should be carefully considered. In this review, we discuss OSCC mouse models and their use in cancer
Mouse model
Genetically modified mouse
research worldwide.
Oral cavity Ó 2017 The Authors. Published by Elsevier Ltd. This is an open access article under the CC BY-NC-ND license
Squamous cell carcinoma (http://creativecommons.org/licenses/by-nc-nd/4.0/).

Introduction which affect genetic regulation [6]. These events are affected by
exposure to environmental agents, including tobacco smoke, alco-
Ninety percent of head and neck cancers are squamous cell car- hol, and viruses (e.g. human papillomavirus), which are the major
cinomas involving mucosal surfaces of the oral cavity, pharynx, risk factors [7,8].
and larynx. This cancer can affect any site in the oral mucosa, typ- Insight into the mechanisms of OSCC initiation and progression
ically the tongue and floor of the mouth [1]. Oral squamous cell is needed, and identification of novel molecular targets to assist the
carcinoma (OSCC), a subset of head and neck squamous cell carci- development of new therapeutic strategies is crucial [9]. New ther-
noma, is one of the most common human malignancies worldwide apies can be investigated both in vitro and in vivo. Animal models,
[2,3]. Further, OSCC is the most common malignant tumor of the particularly mouse models, enable the study of complex biological
head and neck, and its incidence has increased in recent years [4]. processes [10]. An optimal animal model would exhibit sponta-
OSCC is a challenging disease to manage in the field of head and neously occurring oral cancer; however, spontaneous OSCC is
neck cancer. The standard treatment for OSCC is a combination of extremely rare in laboratory animals [11].
surgery, radiation, and chemotherapy. The 5-year survival rate of In mouse models of OSCC, xenograft models and chemical
OSCC is only 50%, which has remained unchanged for a decade carcinogen-induced models are widely used. Xenograft models
[5]. This poor prognosis is considered to stem from aggressive local with implanted human OSCC cells are frequently used to study
invasion and metastasis, leading to recurrence. Further, a lack of human tumor growth and spread as well as to develop and test
applicable markers for early detection and the failure of advanced new antitumor drugs [10].
lesions to respond to chemotherapy contribute to poor OSCC As a chemical carcinogen-induced model, the 4-nitroquinoline
outcomes. 1-oxide (4NQO) mouse model has been used worldwide. The
OSCC lesions accumulate genetic alterations including chromo- dynamics of gene expression alterations in the 4NQO mouse model
somal aberrations, DNA mutations, amplifications, or deletions, of human oral carcinogenesis that lead to the development of pre-
and/or epigenetic alterations, such as changes in DNA methylation, neoplastic histological changes such as hyperplasia and dysplasia
followed by SCC development can be characterized in this model
[12–16].
⇑ Corresponding author.
E-mail address: h_tomita@gifu-u.ac.jp (H. Tomita).

http://dx.doi.org/10.1016/j.oraloncology.2017.07.028
1368-8375/Ó 2017 The Authors. Published by Elsevier Ltd.
This is an open access article under the CC BY-NC-ND license (http://creativecommons.org/licenses/by-nc-nd/4.0/).
K. Ishida et al. / Oral Oncology 73 (2017) 16–20 17

Mice are one of the best model organisms for cancer research in situ (non-invasive SCC), and invasive SCC, even after 4NQO is
because of the animals’ small size, propensity to breed in captivity, withdrawn (Fig. 1) [6,20]. Multiple lesions are considered to consti-
life span of 2–3 years, many physiological and molecular similari- tute the multi-step carcinogenesis of OSCC; this multi-step process
ties to humans, and fully sequenced genome [17]. is one advantage of the 4NQO model, and the development of fully
Here, we review mouse models used to investigate OSCC patho- malignant SCC is preceded by increasing grades of dysplastic
genesis and discuss both the advantages and disadvantages of their changes that mimic oral cancer development in humans. SCCs
application in cancer research. are typically detected between 12 and 33 weeks after applying
4NQO (via drinking water) for 16 weeks [20,21].
Regarding the molecular pathology of the 4-NQO model, the
Chemical carcinogen-induced mouse models SCC tumors produced display some of the molecular changes
observed in human SCC; however, the detailed pathological and
4-Nitroquinolone oxide (4NQO) model molecular mechanisms remain unclear.
The 4NQO molecule forms DNA adducts, causes substitutions of
adenosine for guanosine, and evokes intracellular oxidative stress,
Dimethyl-1,2,benzanthracene (DMBA) model
resulting in genetic mutations and DNA strand breaks. All these
This model involves the administration of polycyclic hydrocar-
effects are similar to the genetic and epigenetic alterations result-
bon 9,10 dimethyl-1,2 benzanthracene (DMBA) dissolved in ben-
ing from exposure to tobacco carcinogens, and therefore, 4NQO
zene or acetone to the cheek pouch of hamsters [22]. DMBA
treatment serves as an alternative for tobacco exposure in animal
induces DNA adduct formation and is mutagenic in mice [23,24].
experiments of oral cancers [13]. 4NQO induces carcinoma in the
However, the DMBA mouse model is not commonly used, and only
tongue and esophagus, and the carcinogenesis model of 4NQO
one study using this model has been reported [25]. In this study,
exposure imitates many situations of human oral squamous
several mice treated with DMBA for 2–6 weeks developed papillo-
carcinogenesis.
mas and papillomas with dysplasia.
Historically, 4NQO, the most commonly used carcinogenic drug,
Overall, carcinogen-induced models do not allow for the study
is a DNA adduct-forming agent that causes DNA damage similar to
of specific genes in the process of oral carcinogenesis. For this pur-
the damage induced by cigarette smoke. In rodent models, the rat
pose, utilizing xenograft models or transgenic mouse models is
oral cavity model was introduced in the early 1970s [18]. In this
necessary.
landmark study, 4NQO dissolved in water produced palatal cancers
in all experimental rats by seven months. In addition, 75% of these
animals developed carcinomas on the dorsum of the tongue and Genetically modified mouse models
20% developed lesions on the gingival tissue [18]. 4NQO has also
been broadly used in mice and is useful for investigating the effects LSL-KrasG12D mouse
of anti-tumor drugs. Another approach involves a novel inducible mouse model that
Regarding mouse models, the 4NQO OSCC model involves a pro- allows oncogene activation and/or tumor suppressor inactivation
tracted multi-step process in which invasive SCC is eventually solely in the stratified epithelia of the oral cavity (Table 1). Two
reached after several months, and 100% of 4NQO-treated mice transgenic mouse models of oral cancer have been described
exhibit precancerous lesions and tumors on the tongues and oral related to K-ras mutations. These models utilize the keratin 5
mucosa [19]. Pathological analyses indicate that these lesions (K5) or keratin 14 (K14) promoter to overexpress the oncogene
and tumors are flat squamous dysplasias (from mild to severe K-rasG12D in the oral epithelium of mice [26,27]. These promoters
atypia), papillary squamous tumors (papillomas), carcinoma are optimal for targeting transgene expression to the oral cavity.

Fig. 1. 4NQO-induced tongue lesions in mice. 4-NQO treatment induces tumors mimicking human oral squamous cell carcinogenesis.
18 K. Ishida et al. / Oral Oncology 73 (2017) 16–20

Table 1 PI3K/Akt pathway. PTEN/PI3K/Akt signaling is thought to play a


Genetically engineerd mouse models of oral squamous cell carcinoma. pivotal role in HNSCC carcinogenesis [31]. TGF-b signaling plays
Model Description Features References both tumor-suppressing and tumor-promoting roles in the car-
LSL-Kras G12D; K5 tet-on-inducible This model develops [26] cinogenesis of various cancer types. In the carcinogenesis of the
K5-rTA form of an papillomas in the Tgfbr1/Pten 2cKO model, PTEN loss leads to hyperproliferation of
oncogenic Kras skin, oral mucosa, the epithelium, and loss of Tgfbr1 contributes to increased inflam-
knock-in tongue, esophagus, or mation, enhanced angiogenesis, and immune suppression. Thus,
forestomach.
LSL-Kras G12D; K5 or K14 Cre- These models [27]
the Tgfbr1/Pten 2cKO model indicates that HNSCC carcinogenesis
K5 or K14 inducible form of develop papillomas, is affected by not only epithelial oncogenic changes, but also the
CrePR1 an oncogenic Kras hyperplasia, andd tumor microenvironment.
knock-in SCCs in the skin, oral
mucosa, tongue,
esophagus, or p53R172H; K5.CrePR1 and p53flox/flox; K5 CrePR1 mice
forestomach. p53 R172H; K5.CrePR1 (16%) and p53 flox/flox; K5 CrePR1 (25%) mice
L2D1+;p53+/ Human CyclinD1 Both models [28,29]
develop oral SCCs [32]. In these models, skin SCCs developed more
and L2D1+; (D1) expression developed invasive
p53 / under the ED-L2 oral-esophageal SCC often than oral SCCs, and metastases were observed. Deleting the
(L2) promoter and Cdkn2a gene enhances skin tumor formation and promotes metas-
p53 homzygous/ tasis in both models; yet, the role of this gene in oral tumors is
heterozygous unclear. p53 R172H; K5.CrePR1 and p53 flox/flox; K5 CrePR1 mouse
knockout
Tgfbr1/Pten K14 Cre-inducible Visible tumors in the [30]
models suggest that both gain and loss of p53 function is associ-
2cKO (Tgfbr1 Tgfbr1 and Pten oral cavity developed ated with HNSCC carcinogenesis.
flox/flox; Pten knockout and the tumors Taken together, the tumor microenvironment in transgenic
flox/flox; K14- histopathologically mice is quite different from that in humans, because mouse stro-
CreER tam) were well
mal cells also contain the transgene. Although the use of oral
differentiated scc
p53 R172H; K5. K5 Cre-inducible In both models, skin [32] mucosa-specific promoters, such as the K5 or K14 promoter, min-
CrePR1 and p53 expression and SCCs developed more imizes the leakage of transgene expression, tissue specificity is not
p53 flox/flox; p53 knockout than oral SCCs, and absolute. Further, a single gene never predominates the natural
K5 CrePR1 observed metastases. process of oral cancer carcinogenesis. The use of one or two more
specific genes such as K-ras or Akt to drive tumor formation in
transgenic mice may not necessarily reflect the carcinogenic pro-
K5 is expressed within the basal epithelium of the tongue and cess in humans. In fact, the frequency of K-ras mutations is low
forestomach, and K14 is mainly expressed in the basal layer of in human head and neck cancer [33,34].
the oral mucosa and tongue [27].
In one model, the K-rasG12D oncogene driven by the K5 or K14
promoter is distributed under the control of Cre recombinase fused Mouse models that combine genetic modification with chemical
to a mutant of the human progesterone receptor [27]. Administra- carcinogen treatment
tion of RU486 induces the overexpression of oncogenic K-rasG12D in
the oral epithelium of mice. Administration of RU486 to mice XPA / ; p53+/ mouse + 4NQO
resulted in the development of squamous papillomas in the oral An XPA / mouse strain carrying mutant alleles for p53 was
cavity. generated to explore the effects of the interaction between nucleo-
In another similar model, expression of the K-rasG12D oncogene tide excision repair machinery with p53 in oral tumorigenesis
driven by the K5 promoter is under the control of tet-responsive (Table 2). In the 4NQO mouse model, despite the same tumor fre-
elements. Oncogenic K-rasG12D overexpression is induced by quency, XPA / p53+/ mice reached 100% SCC incidence at
administering doxycycline to the mice [26]. The doxycycline- 25 weeks compared to 50 weeks for XPA / p53+/+ littermates [35].
inducible mice develop dysplasia and SCC in the skin, oral mucosa,
tongue, esophagus, or forestomach. L2D1+ mouse + 4NQO
The L2D1+ mouse is characterized by human cyclin D1 overex-
L2D1+/p53+/ and L2D1+/p53 / mice pression controlled by the ED2-L2 promoter of the Epstein-Barr
In the head and neck region, the first genetic mouse model virus [28]. Upon 4NQO treatment, this mouse model exhibits accel-
specifically designed to develop SCC was generated by Opitz erated susceptibility to oral dysplasia and SCC [36].
et al. [28]. They used the Epstein-Barr virus ED-L2 promoter (L2)
to target genes specifically in the oral- esophageal squamous PIK3CA-GEMM mouse + 4NQO
epithelium [29] and generated the L2-cyclin D1 (L2D1+) mouse, PIK3CA-GEMM mice are also known as K5. GLp65/tataPIK3CA
which fuses L2 to human cyclin D1 cDNA. They crossed L2D1+ mice mice [37]. This model overexpresses PIK3CA specifically in the buc-
with p53+/ and p53 / mice, resulting in compound mice that cal and tongue epithelium upon RU486 induction. PIK3CA gene
developed invasive oral-esophageal SCC. Further, cell lines from amplification or gain-of-function mutations have been reported
the oral epithelia of L2D1+/p53+/ and L2D1+/p53 / mice induced in human HNSCCs [38–40]. In the PIK3CA-GEMM mouse model,
tumor formation in athymic nu/nu mice [28]. PIK3CA overexpression alone was insufficient for SCC develop-
ment. Upon 4NQO administration, 39% of mice developed SCCs at
Tgfbr1/Pten 2cKO (Tgfbr1flox/flox; Ptenflox/flox; K14-CreERtam) mice 5–6 months and 100% of mice developed SCCs at 10–12 months.
Tgfbr1/Pten 2cKO mice develop hyperplastic changes in the oral This mouse model exhibits enhanced SCC development compared
epithelium 4 weeks after tamoxifen (tam) induction [30]. Ten with wild-type control mice treated with 4NQO. Further, this
weeks after tam induction, visible tumors in the oral cavity mouse model promotes tumor invasion and metastasis into lymph
develop, and histopathology shows that the tumors were well- nodes and lungs. The mechanisms of carcinogenesis in this model
differentiated SCC, mimicking human head and neck SCC (HNSCC). are related to PI3K/PDK1 and TGF-b signaling, similar to the Tgfbr1/
PTEN is a tumor suppressor gene and a negative regulator of the Pten 2cKO mouse model [30].
K. Ishida et al. / Oral Oncology 73 (2017) 16–20 19

Table 2 model of head and neck cancer established from fresh surgical
Mouse models that combine genetic modification with chemical carcinogen specimens of 130 human head and neck cancers implanted subcu-
treatment.
taneously into nude mice.
Model Description Features References One advantage of xenograft models is that several mice injected
XPA / ; p53+/ XPA / knockout 4NQO-treated [5] with oral SCC cells developed lymphatic and pulmonary metasta-
mouse + 4NQO and carrying XPA / p53+/ sis, as observed in human oral SCCs [46,47].
mutant alleles for mice reached 100% One disadvantage of xenograft models is that human cell lines
p53 SCC incidence at
25 weeks
can only be implanted into the tissues of athymic or severe com-
L2D1++ 4NQO Human CyclinD1 By 4NQO, this model [36] bined immunodeficiency mice. The use of immune-deficient mice
(D1) expression accelerated prevents the study of interactions between the tumor and
under the ED-L2 susceptibility to oral microenvironment related to the host immune system.
(L2) promoter dysplasia and SCC
PIK3CA-GEMM PIK3CA-GEMM With 4NQO [37]
mouse + 4NQO mouse is also administration, the
shown as K5. mouse model
GLp65/tataPIK3CA developed SCCs in
Conclusions and future perspectives
mouse. This model 39% mice at 5–
can overexpress 6 months and 100% Mouse models have enabled the use of new approaches to
PIK3CA mice at 10– develop cancer prevention and treatment; identification of early
specifically in the 12 months. Further,
diagnostic markers and novel therapeutic targets; and understand-
buccal and tongue the mouse model
epithelium by promoted tumor ing of the in vivo biology and genetics of tumor initiation, promo-
RU486 induction. invasion and tion, progression, and metastasis. No mouse model is perfectly
metastasis of lymph suitable for studying any type of human cancer. Genetically mod-
node and lung. ified mouse models have only been recently developed and are
K14-GFP-miR-211 K14-GFP-miR-211 By 4NQO [42]
transgenic transgenic mouse administration, this
promising; however, their low tumor penetrance causes uncer-
mouse + 4NQO expresses EGFP mouse model tainty in aspects of experimental design such as endpoint timing
and miR-211 developed SCCs in and actual number of mice available for study [6]. However,
constitutively the tongue, early-stage models would enable the further study of early-stage
driven by K14 esophagus, buccal
indicators (precancerous lesions) and biomarkers, as well as the
promoter in mucosa.
squamous investigation of more appropriate treatments [48].
epithelium Chemical 4NQO carcinogenesis models better reflect the clinical
Dusp1 / mouse Dusp1 / Dusp1 / mice [43] setting because of the diversity and heterogeneous nature of the
+ 4NQO knockout with 4NQO resulting lesions. Chemoprevention has been widely studied using
administration
enhanced oral SCC
4NQO carcinogenesis models. The 4NQO model is attractive for
and tumor- chemoprevention studies because of its easily observable features
associated and tissue sampling for pathologic and molecular analyses.
inflammation with Currently, few neck metastasis models are available for drug tri-
IL1 b upregulation.
als and clinical research on OSCC. In xenograft model, feasible
mouse models of the neck metastasis bone destruction in the jaw
K14-GFP-miR-211 transgenic mouse + 4NQO are developed, and useful for elucidating the metastatic mecha-
MicroRNA-211 (miR-211) promotes HNSCC formation and is nism of OSCC [49,50]. However, the future development of the
implicated in decreased patient survival [41]. K14-GFP-miR-211 genetically modified mouse models with the metastasis could
transgenic mice express EGFP and miR-211 constitutively driven advance the discovery of new therapies for OSCC.
by the K14 promoter in the squamous epithelium [42]. Hyperpro- Animal models in which the early stage of OSCC carcinogenesis
liferation and wall thickness, but not tumor formation, were can be replicated remain to be developed. Such a model will allow
observed in the oral epithelium of these mice. Upon 4NQO admin- for the identification of predictive and correlative biomarkers for
istration, these mice develop SCCs in the tongue, esophagus, and evaluating specific therapeutic approaches.
buccal mucosa. The incidence, number, and diameter of tumors An ideal mouse OSCC model would be a combination of a
in this mouse cohort significantly increases upon 4NQO treatment carcinogen-induced model and transgenic model in which the
compared with tumors in wild type control mice treated with application of carcinogenic agents to transgenic mice leads to early
4NQO. Further, the histology of these tumors mimics the histology tumor formation. This model would be analogous to chronic expo-
of human HNSCCs. sure to tobacco and alcohol in an individual with genetic or epige-
netic predispositions for developing cancers in the oral cavity.
Dusp1 / mouse + 4NQO Until a model resembling human OSCC is developed, both the
Dusp1 / mice exhibit enhanced oral cancer development advantages and disadvantages of each model should be carefully
upon 4NQO administration compared with wild type control mice considered.
[43]. This mouse model exhibits accelerated tumor-associated In conclusion, taking both their advantages and disadvantages
inflammation, including IL1b upregulation. DUSP1 is a dual- into account, OSCC mouse models exhibit clear benefits and appear
specificity phosphatase that controls the mitogen-activated pro- suitable for application in therapeutic research. Clinically relevant
tein kinase (MAPK) pathway [44]. The characteristics of this mouse mouse models that overcome current limitations are required for
model indicate that DUSP1 plays an important role in the tumor studying OSCC chemoprevention and treatment.
microenvironment of oral carcinogenesis.

Transplanted (xenograft) models Acknowledgements

Xenograft models of head and neck cancers have been reported We thank all the members of our laboratory and our collabora-
since the early 1980s. Braakhuis et al. [45] reported a xenograft tors for their research work and helpful discussions.
20 K. Ishida et al. / Oral Oncology 73 (2017) 16–20

This work was partially supported by grants from the Ministry [26] Vitale-Cross L, Amornphimoltham P, Fisher G, Molinolo AA, Gutkind JS.
Conditional expression of K-ras in an epithelial compartment that includes the
of Education, Culture, Sports, Science, and Technology of Japan
stem cells is sufficient to promote squamous cell carcinogenesis. Cancer Res
(#15K11289 and #26430111). 2004;64:8804–7.
[27] Caulin C, Nguyen T, Longley MA, Zhou Z, Wang XJ, Roop DR. Inducible
activation of oncogenic K-ras results in tumor formation in the oral cavity.
Conflicting interests Cancer Res 2004;64:5054–8.
[28] Opitz OG, Harada H, Suliman Y, Rhoades B, Sharpless NE, Kent R, et al. A mouse
The authors declare no conflicts of interest. model of human oral-esophageal cancer. J Clin Invest 2002;110:761–9.
[29] Nakagawa H, Wang TC, Zukerberg L, Odze R, Togawa K, May GH, et al. The
targeting of the cyclin D1 oncogene by an Epstein-Barr virus promoter in
References transgenic mice causes dysplasia in the tongue, esophagus and forestomach.
Oncogene 1997;14:1185–90.
[1] Feller LL, Khammissa RR, Kramer BB, Lemmer JJ. Oral squamous cell carcinoma [30] Bian Y, Hall B, Sun ZJ, Molinolo A, Chen W, Gutkind JS, et al. Loss of TGF-beta
in relation to field precancerisation: pathobiology. Cancer Cell Int 2013;13:31. signaling and PTEN promotes head and neck squamous cell carcinoma through
[2] Siegel RL, Miller KD, Jemal A. Cancer statistics, 2016. CA Cancer J Clin cellular senescence evasion and cancer-related inflammation. Oncogene
2016;66:7–30. 2012;31:3322–32.
[3] Kademani D. Oral cancer. Mayo Clin Proc 2007;82:878–87. [31] Pedrero JM, Carracedo DG, Pinto CM, Zapatero AH, Rodrigo JP, Nieto CS, et al.
[4] Bagan JV, Scully C. Recent advances in Oral Oncology 2007: epidemiology, Frequent genetic and biochemical alterations of the PI 3-K/AKT/PTEN pathway
aetiopathogenesis, diagnosis and prognostication. Oral Oncol 2008;44:103–8. in head and neck squamous cell carcinoma. Int J Cancer 2005;114:242–8.
[5] Brunin F, Mosseri V, Jaulerry C, Point D, Cosset JM, Rodriguez J. Cancer of the [32] Li Z, Gonzalez CL, Wang B, Zhang Y, Mejia O, Katsonis P, et al. Cdkn2a
base of the tongue: past and future. Head Neck 1999;21:751–9. suppresses metastasis in squamous cell carcinomas induced by the gain-of-
[6] Mognetti B, Di Carlo F, Berta GN. Animal models in oral cancer research. Oral function mutant p53(R172H). J Pathol 2016;240:224–34.
Oncol 2006;42:448–60. [33] Munirajan AK, Mohanprasad BK, Shanmugam G, Tsuchida N. Detection of a
[7] Vermorken JB, Mesia R, Rivera F, Remenar E, Kawecki A, Rottey S, et al. rare point mutation at codon 59 and relatively high incidence of H-ras
Platinum-based chemotherapy plus cetuximab in head and neck cancer. N Engl mutation in Indian oral cancer. Int J Oncol 1998;13:971–4.
J Med 2008;359:1116–27. [34] Saranath D, Chang SE, Bhoite LT, Panchal RG, Kerr IB, Mehta AR, et al. High
[8] Vermorken JB, Trigo J, Hitt R, Koralewski P, Diaz-Rubio E, Rolland F, et al. Open- frequency mutation in codons 12 and 61 of H-ras oncogene in chewing
label, uncontrolled, multicenter phase II study to evaluate the efficacy and tobacco-related human oral carcinoma in India. Br J Cancer 1991;63:573–8.
toxicity of cetuximab as a single agent in patients with recurrent and/or [35] Ide F, Kitada M, Sakashita H, Kusama K, Tanaka K, Ishikawa T. P53
metastatic squamous cell carcinoma of the head and neck who failed to haploinsufficiency profoundly accelerates the onset of tongue tumors in
respond to platinum-based therapy. J Clin Oncol 2007;25:2171–7. mice lacking the xeroderma pigmentosum group A gene. Am J Pathol
[9] Ohnishi Y, Inoue H, Furukawa M, Kakudo K, Nozaki M. Heparin-binding 2003;163:1729–33.
epidermal growth factor-like growth factor is a potent regulator of invasion [36] Wilkey JF, Buchberger G, Saucier K, Patel SM, Eisenberg E, Nakagawa H, et al.
activity in oral squamous cell carcinoma. Oncol Rep 2012;27:954–8. Cyclin D1 overexpression increases susceptibility to 4-nitroquinoline-1-oxide-
[10] Szaniszlo P, Fennewald SM, Qiu S, Kantara C, Shilagard T, Vargas G, et al. induced dysplasia and neoplasia in murine squamous oral epithelium. Mol
Temporal characterization of lymphatic metastasis in an orthotopic mouse Carcinog 2009;48:853–61.
model of oral cancer. Head Neck 2014;36:1638–47. [37] Du L, Chen X, Cao Y, Lu L, Zhang F, Bornstein S, et al. Overexpression of PIK3CA
[11] Gardner DG. Spontaneous squamous cell carcinomas of the oral region in in murine head and neck epithelium drives tumor invasion and metastasis
domestic animals: a review and consideration of their relevance to human through PDK1 and enhanced TGFbeta signaling. Oncogene 2016;35:4641–52.
research. Oral Dis 1996;2:148–54. [38] Pickering CR, Zhang J, Yoo SY, Bengtsson L, Moorthy S, Neskey DM, et al.
[12] Tanaka T, Ishigamori R. Understanding carcinogenesis for fighting oral cancer. J Integrative genomic characterization of oral squamous cell carcinoma
Oncol 2011;2011:603740. identifies frequent somatic drivers. Cancer Discov 2013;3:770–81.
[13] Vitale-Cross L, Czerninski R, Amornphimoltham P, Patel V, Molinolo AA, [39] Lui VW, Hedberg ML, Li H, Vangara BS, Pendleton K, Zeng Y, et al. Frequent
Gutkind JS. Chemical carcinogenesis models for evaluating molecular-targeted mutation of the PI3 K pathway in head and neck cancer defines predictive
prevention and treatment of oral cancer. Cancer Prev Res (Phila) biomarkers. Cancer Discov 2013;3:761–9.
2009;2:419–22. [40] Iglesias-Bartolome R, Martin D, Gutkind JS. Exploiting the head and neck
[14] Hasina R, Martin LE, Kasza K, Jones CL, Jalil A, Lingen MW. ABT-510 is an cancer oncogenome: widespread PI3K-mTOR pathway alterations and novel
effective chemopreventive agent in the mouse 4-nitroquinoline 1-oxide model molecular targets. Cancer Discov 2013;3:722–5.
of oral carcinogenesis. Cancer Prev Res (Phila) 2009;2:385–93. [41] Chang KW, Liu CJ, Chu TH, Cheng HW, Hung PS, Hu WY, et al. Association
[15] Baba S, Yamada Y, Hatano Y, Miyazaki Y, Mori H, Shibata T, et al. Global DNA between high miR-211 microRNA expression and the poor prognosis of oral
hypomethylation suppresses squamous carcinogenesis in the tongue and carcinoma. J Dent Res 2008;87:1063–8.
esophagus. Cancer Sci 2009;100:1186–91. [42] Chen YF, Yang CC, Kao SY, Liu CJ, Lin SC, Chang KW. MicroRNA-211 enhances
[16] Kanojia D, Vaidya MM. 4-nitroquinoline-1-oxide induced experimental oral the oncogenicity of carcinogen-induced oral carcinoma by repressing TCF12
carcinogenesis. Oral Oncol 2006;42:655–67. and increasing antioxidant activity. Cancer Res 2016;76:4872–86.
[17] Frese KK, Tuveson DA. Maximizing mouse cancer models. Nat Rev Cancer [43] Zhang X, Hyer JM, Yu H, D’Silva NJ, Kirkwood KL. DUSP1 phosphatase regulates
2007;7:645–58. the proinflammatory milieu in head and neck squamous cell carcinoma.
[18] Wallenius K, Lekholm U. Oral cancer in rats induced by the water-soluble Cancer Res 2014;74:7191–7.
carcinogen 4-nitrochinoline N-oxide. Odontol Revy 1973;24:39–48. [44] Lawan A, Shi H, Gatzke F, Bennett AM. Diversity and specificity of the mitogen-
[19] Foy JP, Tortereau A, Caulin C, Le Texier V, Lavergne E, Thomas E, et al. The activated protein kinase phosphatase-1 functions. Cell Mol Life Sci
dynamics of gene expression changes in a mouse model of oral tumorigenesis 2013;70:223–37.
may help refine prevention and treatment strategies in patients with oral [45] Braakhuis BJ, Sneeuwloper G, Snow GB. The potential of the nude mouse
cancer. Oncotarget 2016. xenograft model for the study of head and neck cancer. Arch Otorhinolaryngol
[20] Tang XH, Knudsen B, Bemis D, Tickoo S, Gudas LJ. Oral cavity and esophageal 1984;239:69–79.
carcinogenesis modeled in carcinogen-treated mice. Clin Cancer Res [46] Myers JN, Holsinger FC, Jasser SA, Bekele BN, Fidler IJ. An orthotopic nude
2004;10:301–13. mouse model of oral tongue squamous cell carcinoma. Clin Cancer Res
[21] Hawkins BL, Heniford BW, Ackermann DM, Leonberger M, Martinez SA, 2002;8:293–8.
Hendler FJ. 4NQO carcinogenesis: a mouse model of oral cavity squamous cell [47] Dinesman A, Haughey B, Gates GA, Aufdemorte T, Von Hoff DD. Development
carcinoma. Head Neck 1994;16:424–32. of a new in vivo model for head and neck cancer. Otolaryngol Head Neck Surg
[22] Salley JJ. Experimental carcinogenesis in the cheek pouch of the Syrian 1990;103:766–74.
hamster. J Dent Res 1954;33:253–62. [48] Schoop RA, Noteborn MH, Baatenburg de Jong RJ. A mouse model for oral
[23] von Pressentin MM, Kosinska W, Guttenplan JB. Mutagenesis induced by oral squamous cell carcinoma. J Mol Histol 2009;40:177–81.
carcinogens in lacZ mouse (MutaMouse) tongue and other oral tissues. [49] Wu TF, Chen L, Bu LL, Gao J, Zhang WF, Jia J. CD44+ cancer cell-induced
Carcinogenesis 1999;20:2167–70. metastasis: a feasible neck metastasis model. Eur J Pharm Sci
[24] Lurie AG, Tatematsu M, Nakatsuka T, Rippey RM, Ito N. Anatomical and 2017;101:243–50.
functional vascular changes in hamster cheek pouch during carcinogenesis [50] Tohyama R, Kayamori K, Sato K, Hamagaki M, Sakamoto K, Yasuda H, et al.
induced by 7, 12-dimethylbenz(a)anthracene. Cancer Res 1983;43:5986–94. Establishment of a xenograft model to explore the mechanism of bone
[25] Kim TW, Chen Q, Shen X, Regezi JA, Ramos DM, Tanaka H, et al. Oral mucosal destruction by human oral cancers and its application to analysis of role of
carcinogenesis in SENCAR mice. Anticancer Res 2002;22:2733–40. RANKL. J Oral Pathol Med 2016;45:356–64.

You might also like