Syngeneic Mouse Models for Therapeutic Research in OC: Comparison
Please note this is a comparison between Version 2 by Conner Chen and Version 1 by Wei-Chiao Chiu.

The most prevalent oral cancer globally is oral squamous cell carcinoma (OSCC). The invasion of adjacent bones and the metastasis to regional lymph nodes often lead to poor prognoses and shortened survival times in patients with OSCC. Encouraging immunotherapeutic responses have been seen with immune checkpoint inhibitors (ICIs); however, these positive responses to monotherapy have been limited to a small subset of patients. Therefore, it is urgent that further investigations into optimizing immunotherapies are conducted. Areas of research include identifying novel immune checkpoints and targets and tailoring treatment programs to meet the needs of individual patients. Furthermore, the advancement of combination therapies against OSCC is also critical. Thus, additional studies are needed to ensure clinical trials are successful. Mice models are advantageous in immunotherapy research with several advantages, such as relatively low costs and high tumor growth success rate. 

  • oral squamous cell carcinoma
  • immune checkpoint inhibitors
  • syngeneic tumor models

1. Introduction

Oral cancer is one of the most common malignant neoplasms in humans, endangering human health, of which more than 90% are oral squamous cell carcinomas (OSCC).
OSCC is a subtype of head and neck squamous cell carcinoma (HNSCC) with an estimated incidence of more than 370,000 new cases and 170,000 deaths annually [1,2,3,4][1][2][3][4]. Globally, the main risks of OSCC are tobacco smoking, alcohol drinking, and betel nut chewing, followed by infection with high-risk human papillomavirus (HPV) [5,6,7,8][5][6][7][8]. OSCC usually occurs in elderly patients; however, the incidence has increased in young people and is mainly due to HPV-associated oropharyngeal squamous cell carcinoma [9]. Over half of OSCC patients are diagnosed at the T3 or T4 stage of disease progression, during which cancer invades local bones of the maxilla or mandible or metastasizes to regional lymph nodes [10]. In addition to lymph node metastasis, local infiltration of submucosa and bone is a common histological feature of OSCC [11,12][11][12]. Different types of bone invasion, including erosive, infiltrative, and mixed patterns, have been found in OSCC, which have different histological features and 3-year disease-free incidence. Compared with the erosive bone invasion pattern of OSCC, infiltrative bone invasive OSCC has a lower rate of 3-year disease-free status [13].
So far, despite the advanced technology in surgery, chemotherapy, and radiotherapy, the survival rate has hardly improved in the past two decades. The tumor microenvironment (TME) contains many different normal cells that have an essential role in tumor development and progression. The stromal fibroblasts, extracellular matrix, blood vessels, lymphatic vessels, infiltrating immune cells, growth factors, and cytokines secreted by TME cells all have positive and negative effects on tumor development [14,15,16,17,18][14][15][16][17][18].
Immunotherapy was developed through advances in knowledge of the interaction between the immune system and tumors and has improved treatment prospects in cancer patients. The methods of immunotherapy assist the immune components in the TME to resist the ability of the tumor to escape immune surveillance, by which the innate immune cells eliminate cancer cells or enhance the anti-tumor immune response [19]. The immune checkpoint blockade (ICB) approach, one of the immunotherapies, aims to drive the immune system to generate an effective anti-tumor response [20,21][20][21]. Immune checkpoint inhibitors (ICIs) are a new kind of anti-tumor immunotherapeutic agent that can inhibit many immune checkpoints, especially on cytotoxic T cells [22,23][22][23]. Identifying novel immune checkpoints and targets and tailoring treatment to individual patients is one focus area in immunotherapy. The immunotherapeutic effects of immune checkpoint inhibitors have been encouraging; however, only a limited subset of patients respond to monotherapy. Therefore, it is urgent to carry out further research, develop new combination therapies, develop more immunotherapeutic drugs, and improve the success rate of clinical trials.
To evaluate the effectiveness of immunotherapy, animal models need to be established in which human tumors and their microenvironment are genetically, physiologically, and anatomically modeled in order to faithfully reflect the formation and development of human tumors. Compared with other animal models, mice have lower costs, shorter reproductive cycles, higher tumor growth rates, and easy genetic modification. Furthermore, established inbred mice allow for tumor transplantation among the same strain of mice or cell lines from the same strain of mice. These advantages make the mouse model a good tool for evaluating the effectiveness of cancer immunotherapy. However, the ability to transfer encouraging immunotherapy results from preclinical trials to the clinic is now a challenge because, after promising results in mouse models, high failure rates have been observed in human clinical trials [24,25][24][25].
Oral carcinogenesis is a complex process composed of oral carcinogens (i.e., alcohol, tobacco, betel nut) and/or human HPV caused by a variety of genetic and epigenetic changes. The tumor microenvironments in mouse models that mimic the human cancer growth genetically, physiologically, and anatomically are important for the research of OSCC immunotherapy [24,26,27][24][26][27]. While a single model cannot recapitulate all aspects of OSCC, the data gathered from animal models are vital for advancing OSCC diagnosis and treatment [28,29][28][29].

2. Syngeneic Mouse Models

Syngeneic OSCC mouse models are produced using allografts of immortalized mouse tumor cell lines. The models can efficiently prevent tissue rejection or graft-versus-host disease (GvHD), which results from transplanting tumor cells into mice. In addition, these models have the inherent advantage of fast establishment, high stability, and high consistency of transplanted tumors. Therefore, they are widely used in immuno-oncology studies and show great potential in developing novel OSCC treatments, especially immunotherapy [70][30]. Tumor cells can be injected orthotopically or ectopically. The orthotopic model supplies a more exact tumor microenvironment, while the subcutaneous model facilitates tumor monitoring and handling. For oral cancer induction, orthotopic models are performed by injecting tumor cells into the oral cavity, while ectopic models most commonly receive subcutaneous injections in the flanks [24]. The principle of syngeneic models is similar to the cell-derived xenograft (CDX) model. Several mouse cell lines can be used to develop syngeneic OSCC mouse models, including mouse OSCC Sq-1979 cells [36][31], mouse squamous cell carcinoma SCC7 [37[32][33],38], mouse oral cancer (MOC) cell lines [40][34], MOC1 [41][35], and MOC2 [42][36]. Moroishi et al. [39][37] have demonstrated that transplanting SCC7 cells (1 × 105) into both hind flanks of C3H/HeOu mice resulted in aggressive tumor growth, whereas LATS1/2 dKO SCC7 cells did not result in tumors. Similarly, Dong et al. [38][33] injected SCC7 cells (1 × 106) into the abdomen of C3H/HeJ mice to assess the effectiveness of a tumor-derived autophagosome vaccine (DRibble). Nagaya et al. [40][34] studied the effects of near-infrared photoimmunotherapy using syngeneic models developed through the subcutaneous injection of C57BL/6 mice with poorly immunogenic MOC2 mKate2 cells (1.5 × 105), moderately immunogenic MOC2-luc cells (1.5 × 105), and immunogenic MOC1 cells (2.0 × 106). Similarly, Adachi et al. [36][31] injected Sq-1979 cells (1 × 107) into the posterior neck area of C3H/HeN mice to determine the genetic changes that occur throughout OSCC development. These studies demonstrate that different mouse OSCC cell lines could be successfully used to produce stable syngeneic OSCC models. In addition, several researchers use 4-nitroquinoline-1-oxide (4NQO) to induce OSCC in a mouse and then inject the OSCC obtained from the mouse into another mouse of the same species to establish a syngeneic model. This is similar to the construction of patient-derived xenograft (PDX) models. For example, Chen et al. used 4NQO (100 μg/mL in drinking water) to induce C57BL/6 mice over 16 weeks; the mice were sacrificed at week 28 to generate the mouse tongue OSCC cell lines MTCQ1 and MTCQ2. Afterward, the MTCQ1 or MTCQ2 cells were injected into the flank or tongue of new mice to establish ectopic or orthotopic mouse models, respectively [43][38]. Compared with human SAS tongue SCC cell lines, MTCQ cells have lower proliferation ability but far higher abilities of migration/invasion. Such capabilities are demonstrated through the identification of extensive cervical lymph node metastasis and lung metastasis resulting from an MTCQ1 cell subclone. Several therapeutic approaches have been tested using this model, including anti-PD-L1 immunotherapy, cisplatin therapy, and miRNAs (especially miR-134). In addition, Chen et al. [44][39] established a syngeneic model using 4NQO-induced OSCC transgenic mice. In this model, K14-EGFP-miR-211 transgenic mice were induced using 4NQO (100 μg/mL in drinking water) over 16 weeks and then sacrificed. Tissue isolated from OSCC lesions on the dorsal surface of the tongue was then used to produce cell lines MOC-L1 to -L4. These were subsequently used to create orthotopic xenografts and real-time in vivo tumor imaging by injecting cells (5 × 106) into the central tongue of C57BL/6 mice. These cells were also used to measure the efficacy of cisplatin therapy and study distant metastasis. Chen et al. [45][40] established the NHRI-HN1 and NHRI-HN2 cell lines from 4-NQO/arecoline-induced murine tongue tumors and further selected for cell stemness by in vitro sphere culture to evaluate potential immunotherapy for OSCC in East and Southeast Asia. NHRI-HN1 or NHRI-HN2 cells (5 × 105) in 50 μL in sterile phosphate-buffered saline (PBS) were injected into the buccal mucosa of mice that were sacrificed 40 days after injection. Syngeneic tumor models have also been applied to the investigation of the anti-tumor activity of ICIs, including anti-programmed death (PD)-1/anti-PD-ligand 1 (L1) antibodies [71,72][41][42] and anti-cytotoxic T lymphocyte-associated antigen 4 (CTLA-4) [46][43]. The time taken to produce syngeneic tumor models is short, as tumor growth happens within a few weeks [25,73][25][44]. However, such rapid tumor growth can prevent the assessment of immunotherapeutics, as the treatment effect is often progressive and estimated by improving survival [74][45]. This makes syngeneic models unsuitable for assessing immunotherapy drugs at the early stages of tumor development [75][46]. The syngeneic OSCC mouse model is a viable tool for immuno-oncology. Still, the main problem is that the model only represents mouse oral cancer and forms mouse tumors with mouse targets. Mice and humans differ in compositions and mechanisms, and some targets in humans are absent or unresponsive in mice.

References

  1. Park, J.; Zhang, X.; Lee, S.K.; Song, N.-Y.; Son, S.H.; Kim, K.R.; Shim, J.H.; Park, K.-K.; Chung, W.-Y. CCL28-induced RARβ expression inhibits oral squamous cell carcinoma bone invasion. J. Clin. Investig. 2019, 129, 5381–5399.
  2. Fan, H.; Tian, H.; Cheng, X.; Chen, Y.; Liang, S.; Zhang, Z.; Liao, Y.; Xu, P. Aberrant Kank1 expression regulates YAP to promote apoptosis and inhibit proliferation in OSCC. J. Cell. Physiol. 2020, 235, 1850–1865.
  3. Sung, H.; Ferlay, J.; Siegel, R.L.; Laversanne, M.; Soerjomataram, I.; Jemal, A.; Bray, F. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J. Clin. 2021, 71, 209–249.
  4. Ali, K. Oral cancer-the fight must go on against all odds... Evid.-Based Dent. 2022, 23, 4–5.
  5. Bandhary, S.K.; Shetty, V.; Saldanha, M.; Gatti, P.; Devegowda, D.; R, P.S.; Shetty, A.K. Detection of Human Papilloma Virus and Risk Factors among Patients with Head and Neck Squamous Cell Carcinoma Attending a Tertiary Referral Centre in South India. Asian Pac. J. Cancer Prev. 2018, 19, 1325–1330.
  6. Auguste, A.; Deloumeaux, J.; Joachim, C.; Gaete, S.; Michineau, L.; Herrmann-Storck, C.; Duflo, S.; Luce, D. Joint effect of tobacco, alcohol, and oral HPV infection on head and neck cancer risk in the French West Indies. Cancer Med. 2020, 9, 6854–6863.
  7. Ahmad, P.; Nawaz, R.; Qurban, M.; Shaikh, G.M.; Mohamed, R.N.; Nagarajappa, A.K.; Asif, J.A.; Alam, M.K. Risk factors associated with the mortality rate of oral squamous cell carcinoma patients: A 10-year retrospective study. Medicine 2021, 100, e27127.
  8. Aghiorghiesei, O.; Zanoaga, O.; Nutu, A.; Braicu, C.; Campian, R.S.; Lucaciu, O.; Berindan Neagoe, I. The World of Oral Cancer and Its Risk Factors Viewed from the Aspect of MicroRNA Expression Patterns. Genes 2022, 13, 594.
  9. Zygogianni, A.G.; Kyrgias, G.; Karakitsos, P.; Psyrri, A.; Kouvaris, J.; Kelekis, N.; Kouloulias, V. Oral squamous cell cancer: Early detection and the role of alcohol and smoking. Head Neck Oncol. 2011, 3, 2.
  10. Koyfman, S.A.; Ismaila, N.; Crook, D.; D’Cruz, A.; Rodriguez, C.P.; Sher, D.J.; Silbermins, D.; Sturgis, E.M.; Tsue, T.T.; Weiss, J.; et al. Management of the Neck in Squamous Cell Carcinoma of the Oral Cavity and Oropharynx: ASCO Clinical Practice Guideline. J. Clin. Oncol. 2019, 37, 1753–1774.
  11. Ukpo, O.C.; Flanagan, J.J.; Ma, X.-J.; Luo, Y.; Thorstad, W.L.; Lewis, J.S. High-Risk Human Papillomavirus E6/E7 mRNA Detection by a Novel In Situ Hybridization Assay Strongly Correlates With p16 Expression and Patient Outcomes in Oropharyngeal Squamous Cell Carcinoma. Am. J. Surg. Pathol. 2011, 35, 1343–1350.
  12. Smith, L.P.; Thomas, G.R. Animal models for the study of squamous cell carcinoma of the upper aerodigestive tract: A historical perspective with review of their utility and limitations. Part A. Chemically-induced de novo cancer, syngeneic animal models of HNSCC, animal models of transplanted xenogeneic human tumors. Int. J. Cancer 2006, 118, 2111–2122.
  13. Simmons, J.K.; Hildreth, B.E., 3rd; Supsavhad, W.; Elshafae, S.M.; Hassan, B.B.; Dirksen, W.P.; Toribio, R.E.; Rosol, T.J. Animal Models of Bone Metastasis. Vet. Pathol. 2015, 52, 827–841.
  14. Kitaeva, K.V.; Rutland, C.S.; Rizvanov, A.A.; Solovyeva, V.V. Cell Culture Based in vitro Test Systems for Anticancer Drug Screening. Front. Bioeng. Biotechnol. 2020, 8, 322.
  15. Chen, Y.-J.; Chang, J.T.-C.; Liao, C.-T.; Wang, H.-M.; Yen, T.-C.; Chiu, C.-C.; Lu, Y.-C.; Li, H.-F.; Cheng, A.-J. Head and neck cancer in the betel quid chewing area: Recent advances in molecular carcinogenesis. Cancer Sci. 2008, 99, 1507–1514.
  16. Mun, J.-Y.; Leem, S.-H.; Lee, J.H.; Kim, H.S. Dual Relationship Between Stromal Cells and Immune Cells in the Tumor Microenvironment. Front. Immunol. 2022, 13, 864739.
  17. Lv, C.; Li, S.; Zhao, J.; Yang, P.; Yang, C. M1 Macrophages Enhance Survival and Invasion of Oral Squamous Cell Carcinoma by Inducing GDF15-Mediated ErbB2 Phosphorylation. ACS Omega 2022, 7, 11405–11414.
  18. Hadjigol, S.; Shah, B.; O’Brien-Simpson, N. The ‘Danse Macabre’—Neutrophils the Interactive Partner Affecting Oral Cancer Outcomes. Front. Immunol. 2022, 13, 894021.
  19. Chulpanova, D.S.; Kitaeva, K.V.; Green, A.R.; Rizvanov, A.A.; Solovyeva, V.V. Molecular Aspects and Future Perspectives of Cytokine-Based Anti-cancer Immunotherapy. Front. Cell Dev. Biol. 2020, 8, 402.
  20. Zou, W.; Wolchok, J.D.; Chen, L. PD-L1 (B7-H1) and PD-1 pathway blockade for cancer therapy: Mechanisms, response biomarkers, and combinations. Sci. Transl. Med. 2016, 8, 328rv324.
  21. Ribatti, D. The concept of immune surveillance against tumors. The first theories. Oncotarget 2017, 8, 7175–7180.
  22. Marin-Acevedo, J.A.; Dholaria, B.; Soyano, A.E.; Knutson, K.L.; Chumsri, S.; Lou, Y. Next generation of immune checkpoint therapy in cancer: New developments and challenges. J. Hematol. Oncol. 2018, 11, 39.
  23. Naimi, A.; Mohammed, R.N.; Raji, A.; Chupradit, S.; Yumashev, A.V.; Suksatan, W.; Shalaby, M.N.; Thangavelu, L.; Kamrava, S.; Shomali, N.; et al. Tumor immunotherapies by immune checkpoint inhibitors (ICIs); the pros and cons. Cell Commun. Signal. 2022, 20, 44.
  24. Li, E.; Lin, L.; Chen, C.-W.; Ou, D.-L. Mouse Models for Immunotherapy in Hepatocellular Carcinoma. Cancers 2019, 11, 1800.
  25. Chulpanova, D.S.; Kitaeva, K.V.; Rutland, C.S.; Rizvanov, A.A.; Solovyeva, V.V. Mouse Tumor Models for Advanced Cancer Immunotherapy. Int. J. Mol. Sci. 2020, 21, 4118.
  26. Luo, J.J.; Young, C.D.; Zhou, H.M.; Wang, X.J. Mouse Models for Studying Oral Cancer: Impact in the Era of Cancer Immunotherapy. J. Dent. Res. 2018, 97, 683–690.
  27. Johnson, D.E.; Burtness, B.; Leemans, C.R.; Lui, V.W.Y.; Bauman, J.E.; Grandis, J.R. Head and neck squamous cell carcinoma. Nat. Rev. Dis. Primers 2020, 6, 92.
  28. Ballegeer, E.A.; Madrill, N.J.; Berger, K.L.; Agnew, D.W.; McNiel, E.A. Evaluation of hypoxia in a feline model of head and neck cancer using 64 Cu-ATSM positron emission tomography/computed tomography. BMC cancer 2013, 13, 218.
  29. Wypij, J.M. A naturally occurring feline model of head and neck squamous cell carcinoma. Pathol. Res. Int. 2013, 2013, 502197.
  30. Li, Q.; Dong, H.; Yang, G.; Song, Y.; Mou, Y.; Ni, Y. Mouse Tumor-Bearing Models as Preclinical Study Platforms for Oral Squamous Cell Carcinoma. Front. Oncol. 2020, 10, 212.
  31. Adachi, M.; Mizuno-Kamiya, M.; Takayama, E.; Kawaki, H.; Inagaki, T.; Sumi, S.; Motohashi, M.; Muramatsu, Y.; Sumitomo, S.I.; Shikimori, M.; et al. Gene expression analyses associated with malignant phenotypes of metastatic sub-clones derived from a mouse oral squamous cell carcinoma Sq-1979 cell line. Oncol. Lett. 2018, 15, 3350–3356.
  32. Su, H.; Luo, Q.; Xie, H.; Huang, X.; Ni, Y.; Mou, Y.; Hu, Q. Therapeutic antitumor efficacy of tumor-derived autophagosome (DRibble) vaccine on head and neck cancer. Int. J. Nanomed. 2015, 10, 1921.
  33. Dong, H.; Su, H.; Chen, L.; Liu, K.; Hu, H.-m.; Yang, W.; Mou, Y. Immunocompetence and mechanism of the DRibble-DCs vaccine for oral squamous cell carcinoma. Cancer Manag. Res. 2018, 10, 493.
  34. Nagaya, T.; Nakamura, Y.; Okuyama, S.; Ogata, F.; Maruoka, Y.; Choyke, P.L.; Allen, C.; Kobayashi, H. Syngeneic Mouse Models of Oral Cancer Are Effectively Targeted by Anti-CD44-Based NIR-PIT. Mol. Cancer Res. 2017, 15, 1667–1677.
  35. Chung, M.K.; Jung, Y.H.; Lee, J.K.; Cho, S.Y.; Murillo-Sauca, O.; Uppaluri, R.; Shin, J.H.; Sunwoo, J.B. CD271 Confers an Invasive and Metastatic Phenotype of Head and Neck Squamous Cell Carcinoma through the Upregulation of Slug. Clin. Cancer Res. 2018, 24, 674.
  36. Judd, N.P.; Allen, C.T.; Winkler, A.E.; Uppaluri, R. Comparative Analysis of Tumor-Infiltrating Lymphocytes in a Syngeneic Mouse Model of Oral Cancer. Otolaryngol.–Head Neck Surg. 2012, 147, 493–500.
  37. Moroishi, T.; Hayashi, T.; Pan, W.-W.; Fujita, Y.; Holt, M.V.; Qin, J.; Carson, D.A.; Guan, K.-L. The Hippo pathway kinases LATS1/2 suppress cancer immunity. Cell 2016, 167, 1525–1539.e1517.
  38. Chen, Y.-F.; Chang, K.-W.; Yang, I.T.; Tu, H.-F.; Lin, S.-C. Establishment of syngeneic murine model for oral cancer therapy. Oral Oncol. 2019, 95, 194–201.
  39. Chen, Y.-F.; Liu, C.-J.; Lin, L.-H.; Chou, C.-H.; Yeh, L.-Y.; Lin, S.-C.; Chang, K.-W. Establishing of mouse oral carcinoma cell lines derived from transgenic mice and their use as syngeneic tumorigenesis models. BMC Cancer 2019, 19, 281.
  40. Chen, Y.-L.; Liu, K.-J.; Jang, C.-W.; Hsu, C.-C.; Yen, Y.-C.; Liu, Y.-L.; Chuang, T.-H.; Wang, S.-H.; Fu, Y.-K.; Kuo, C.-C.; et al. ERK Activation Modulates Cancer Stemness and Motility of a Novel Mouse Oral Squamous Cell Carcinoma Cell Line. Cancers 2019, 12, 61.
  41. Kim, S.-S.; Harford, J.B.; Moghe, M.; Slaughter, T.; Doherty, C.; Chang, E.H. A tumor-targeting nanomedicine carrying the p53 gene crosses the blood–brain barrier and enhances anti-PD-1 immunotherapy in mouse models of glioblastoma. Int. J. Cancer 2019, 145, 2535–2546.
  42. Jiao, R.; Allen, K.J.H.; Malo, M.E.; Rickles, D.; Dadachova, E. Evaluating the Combination of Radioimmunotherapy and Immunotherapy in a Melanoma Mouse Model. Int. J. Mol. Sci. 2020, 21, 773.
  43. Wang, Z.; Wu, V.H.; Allevato, M.M.; Gilardi, M.; He, Y.; Luis Callejas-Valera, J.; Vitale-Cross, L.; Martin, D.; Amornphimoltham, P.; McDermott, J.; et al. Syngeneic animal models of tobacco-associated oral cancer reveal the activity of in situ anti-CTLA-4. Nat. Commun. 2019, 10, 5546.
  44. Ngiow, S.F.; Loi, S.; Thomas, D.; Smyth, M.J. Mouse Models of Tumor Immunotherapy. Adv. Immunol. 2016, 130, 1–24.
  45. Olson, B.; Li, Y.; Lin, Y.; Liu, E.T.; Patnaik, A. Mouse Models for Cancer Immunotherapy Research. Cancer Discov. 2018, 8, 1358–1365.
  46. Gulley, J.L.; Drake, C.G. Immunotherapy for prostate cancer: Recent advances, lessons learned, and areas for further research. Clin. Cancer Res. 2011, 17, 3884–3891.
More