• 沒有找到結果。

5.2.1 Significance

We demonstrated in this study the in vitro and in vivo radiosensitising effect of afatinib

(an irreversible EGFR/HER2 dual inhibitor) on MBT-2 murine bladder cancer model. The

synergism with irradiation was likely mediated through enhancement of radiation-induced

DNA damage and apoptosis. We also showed that afatinib suppressed radiation-activated

EGFR and HER2 signaling. To our knowledge, the present study is the first to report that the

combined blockade of EGFR and other Erb-B family receptor tyrosine kinases enhances the

radiosensitivity of bladder cancer cells.

EGFR specific tyrosine kinase inhibitor like gefitinib (Colquhoun et al., 2007;

Dominguez-Escrig et al., 2004; Maddineni, Sangar, Hendry, Margison, & Clarke, 2005) and

erlotinib (Colquhoun & Mellon, 2004) has been reported to enhance the radiosensitivity of

bladder cancer cells. Compared to the gefitinib, the radiosensitizing effect of afatinib in our

study was superior, indicating the critical role of HER2 signaling pathway in determining the

radiosensitivity of bladder cancer cells. Similarly, monoclonal antibody to HER2/neu receptor

has been shown to modulate the repair of radiation-induced DNA damage and enhance the

radiosensitivity of breast and esophageal cancer cells overexpressing HER2 (Pietras et al.,

1999; Sato et al., 2005). Furthermore, the Radiation Therapy Oncology Group trial on

bladder preservation demonstrated a significant association of HER2 staining of bladder

tumor tissue with complete response after tri-modality treatment (Chakravarti et al., 2005).

Lapatinib, a reversible EGFR/HER2 dual inhibitor, had been shown to have a

radiosensitizing effect in certain breast cancer cells (Sambade, Camp, Kimple, Sartor, &

Shields, 2009; Sambade et al., 2010) and patients (Abboud, Saghir, Salame, & Geara, 2010;

Harrington et al., 2009). However, its effect was unsatisfactory in our pilot study. Compared

to lapatinib, afatinib (an irreversible inhibitor of EGFR and HER2) was the more potent

radiosensitizer (Li et al., 2008). One large randomized trial recently reported the clinical

benefit of afatinib for patients with non-small-cell lung cancer unresponsive to EGFR

inhibitors or chemotherapy (Miller et al., 2012). Furthermore, afatinib had been shown to

enhance responses to an EGFR specific monoclonal antibody in bladder cancer cells resistant

to this treatment (Quesnelle & Grandis, 2011).

Schütze et al. investigated radiosensitization by afatinib of human squamous cell

carcinoma cells only marginally affected by radiation (Abraham, Pagano, Gomella, & Baffa,

2007). The design of the present study differed from their design in the in vivo dosing

schedule of afatinib (10 mg/kg for 7 days in our study versus 20 mg/kg until reaching a tumor

diameter of 15 mm in their study) and radiotherapy (15 Gy on day 4 in this study versus 20

Gy after the last afatinib dose in their study). Moreover, we measured not only the volume of

the ectopic tumor but also its glucose metabolism by PET, as well as the corresponding

expression of EGFR and HER2 immunohistochemically. We additionally showed that

afatinib enhanced radiation-induced DNA damage and apoptosis. The molecular response to

irradiation combined with afatinib might vary between different malignant cells (Krause,

Gurtner, Deuse, & Baumann, 2009; Toulany, Dittmann, Baumann, & Rodemann, 2005) and

this variation might account for discrepancies between our study and the one by Schutze et al.

MBT-2 cells are poorly differentiated urothelial carcinoma and derived from a

carcinogen-induced bladder tumor in C3H/He mice. The tumor resembles its human

counterpart both grossly and histologically (Horiguchi et al., 2008). The advantage of the

MBT-2 murine bladder cancer model in this study is the immune-competent system for the in

vivo data with the integrated physiological response after irradiation (Blomgren, Edsmyr, von

Stedingk, & Wasserman, 1986) and inhibition of Erb-B family receptor tyrosine kinases

(Hamilton et al., 2012; Y. Yan et al., 2006).

In this study, radiosensitization by afatinib was mediated through enhanced

irradiation-induced DNA damage as indicated by the increase in γ-H2AX foci. γ-H2AX is a marker of

DNA double-strand breaks (Rogakou, Pilch, Orr, Ivanova, & Bonner, 1998). Increased numbers of γ-H2AX foci have been found in bronchial carcinoma and breast adenoma cells

treated with a EGFR inhibitor for radiosensitization (Dittmann, Mayer, & Rodemann, 2005),

and this increase has been correlated with dead or dying cells (Gonzalez, Barquinero, Lee,

Garcia, & Casaco, 2012). Double-strand breaks of DNA are the principal lesions responsible

for cell killing by ionizing radiation. The radiosensitivity and clonogenic survival of

irradiated bladder cancer cell lines have been previously assessed by alkaline comet assay, a

method to detect DNA strand break damage (Moneef et al., 2003). Our findings of reduced clonogenic survival and significantly increased numbers of γ-H2AX foci after combined

afatinib and irradiation imply that afatinib augments DNA damage. Given that DNA is the

most important cellular target of ionizing radiation, afatinib is an ideal radiosensitizer for

bladder cancer.

5.2.2 Limitation

Although we demonstrate that afatinib suppresses irradiation-activated EGFR and HER2

pathways, the full mechanism of enhanced radiosensitization of bladder cancer cells remains

elusive. Sambade et al. reported that lapatinib mediates radiosensitization in breast cancer

cells primarily by inhibiting the Raf > MEK > ERK mitogen-activated protein kinase cascade

(Sambade et al., 2009). We showed that afatinib inhibits post-radiation Akt phosphorylation

in response to EGFR and HER2 signaling. Nicolle et al. similarly found that growth and

invasiveness inhibition by gefitinib in urothelial carcinoma cell lines involves strong

phosphorylation of Akt/MAPK pathways in association with activation of EGFR (Nicolle et

al., 2006). Further investigation is required to identify the pathways through which DNA

damage and apoptosis signals are transduced.

5.2.3 Conclusion

In this murine bladder cancer model we demonstrated for the first time the in vitro and

in vivo radiosensitizing activity of afatinib, an orally bioavailable EGFR/HER2 dual inhibitor.

Afatinib likely mediates its effect on bladder cancer cells by suppressing radiation-activated

EGFR and HER2 signals and thereby causing enhanced DNA damage and cell apoptosis. The

greater potency of afatinib as a radiosensitizer compared with previously reported EGFR

inhibitors underscores the importance of other Erb-B family receptor tyrosine kinases such as

HER2, and indicates a new direction for future clinical trials in bladder cancer.

相關文件