• 沒有找到結果。

Unfinished Study: Mutant EGFR - Related Research

5.4.1 Planned experiments

We have proved that both T24 and NTUB1 human bladder cancer cells have wild-type

EGFR. On the other hand, non-small cell lung cancer with the L858R or the ΔE746-E750

mutations in the tyrosine kinase domain of EGFR was reported to exhibit enhanced

sensitivity to radiation, and this strongly correlated with dramatically diminished capacity to

resolve radiation-induced DNA double strand breaks (Das et al., 2006). Moreover, non-small

cell lung cancer patients with mutant EGFR patients treated with radiotherapy had a better

clinical outcome than patients with wild-type EGFR in terms of locoregional control (Mak et

al., 2011) or response to whole brain radiotherapy (Gow et al., 2008). It is also reported that

the mutant EGFR in these non-small cell lung carcinomas failed to exhibit radiation-induced

nuclear translocation or binding to DNA-PKcs (Das et al., 2007). Therefore it is plausible to

hypothesize that EGFR mutations may confer sensitivity to radiation.

To introduce different forms of EGFRs into T24 cells, we used the pLenti6/directional

TOPO cloning kit. Full-length fragment of wild-type EGFR was amplified from

pcDNA3.1-EGFR-wt and cloned into pLenti6/directional TOPO vector according to the instructions of

the manufacturer. The L858R mutation was introduced by using site-directed mutagenesis kit

(Stratagene). We also planned to introduce the full length of E746-A750 del mutation from

cDNA of HCC827 cell line and cloned into pLenti6/directional TOPO vector. Correct

sequences were confirmed by sequencing for all vectors. However, although we successfully

introduced the plasmid with mutated EGFR to T24 bladder cancer cell, the majority of EGFR

in T24 cells was the wild-type. After evaluation we abandoned the rest of the experiments.

5.4.2 Lessons learned from recent literature

Interestingly, Zhang et al. recently showed that afatinib increases radiosensitivity of

non-small cell lung cancer cells with acquired EGFR T790M mutation (Zhang et al., 2015). Since

the experimental design is similar to ours, it deserves description and comparison. In Fig. 5-1

the authors showed the change of EGFR/Akt/ERK pathways in lung cancer cells after

irradiation and afatinib. The phosphorylation levels of EGFR, Akt and ERK increased after

irradiation in PC-9 (EGFR del E746-A750), PC-9-GR (PC-9 with gefitinib resistance) and

H1975 cells (EGFR L858R and T790M mutation). Pretreatment with afatinib remarkable

blocked basal level of the phosphorylations of EGFR and ERK proteins, and caused delays of

irradiation-induced phosphorylation of Akt in these cells.

Figure 5-1. Effects of afatinib on protein phosphorylation after irradiation or afatinib

pretreatment+irradiation in PC-9 (A), PC-9-GR (B) and H1975 (C) cells (Zhang et al., 2015)

But afatinib did not cause changes of the basal levels for phosphorated EGFR, Akt and

ERK proteins in H460 (wild-type EGFR) cells which has a low baseline of these proteins

(Fig. 5-2). Although we don’t have the data about EGFR-mutated bladder cancer cells, the

result confirmed our previous observation in Fig. 3-2, Fig 3-3 and Fig 4-1 that afatinib

inhibits certain radiation-activated signaling pathways.

Figure 5-2. The effects of afatinib on protein phosphorylations in lung cancer cells (Zhang et

al., 2015)

The authors also demonstrated the clonogenic assay in PC-9, PC-9-GR, H1975 and

H460 cells after the treatment of radiation and/or afatinib (Fig. 5-3). PC-9-GR cells which

acquired T790M mutation in addition to the original deletion in exon 19 (del E746-A750)

demonstrated the radiosensitizing effect of afatinib. Similar to our experimental design, the

authors showed that afatinib treatment lead to increased apoptosis (by flow cytometry) and

suppressed DNA damage repair (by γ-H2AX foci) in irradiated PC-9-GR cells, and enhanced

tumor growth inhibition when combined with irradiation in PC-9-GR xenografts. However,

the exact mechanism why lung cancer cells with different EGFR mutation have different

susceptibility to afatinib-induced radiosensitization was not addressed

Figure 5-3. Effect of afatinib on clonogenic survival in irradiated lung cancer cells

(Zhang et al., 2015)

It is also a pity that the expression of baseline and radiation-induced HER2

phosphorylation was not compared in this study. Although H460 cell as well as T24 and

NTUB1 bladder cancer cells in our study has wild-type EGFR, the extent of HER2 activation

after irradiation may explain the difference in radiosensitizing effect of afatinib. More

mechanistic studies are still awaited.

CHAPTER SIX: PROSPECT

6.1 Radiosensitizing Activity of Afatinib and Microenvironment

Till now our study of the radiosensitizing effects of new-generation EGFR inhibitor

focus on the factors influence cell growth, like DNA damage and apoptosis. On the other

hand, the interaction between cancer cells and microenvironment also determine the potential

to invasion and metastasis, therefore it is very important to the radiation effect. Rapidly

accumulating evidence suggests that radiation exposure also promotes cancer metastatic

ability through epithelial-mesenchymal transition (EMT), which has a central role in cancer

metastasis and has become the subject of intense investigation (Kawamoto et al., 2012; Liu et

al., 2014; S. Yan et al., 2013). However, the signaling molecular mechanisms underlying

radiation-induced EMT remain obscure (Cui et al., 2015), and there is virtually no report

regarding this field in bladder cancer research. Since the failure pattern of radiotherapy in

bladder cancer patients includes local recurrence and distant metastasis, the influence of how

new-generation EGFR TKIs like afatinib on these phenomena will be of great significance.

Till now we have some preliminary result. As shown in Fig. 6-1, Boyden chamber

invasion assay revealed that after 2.5Gy irradiation, there is an increase of cell invasion in

T24 and 5637 bladder cancer cells. After pretreatment with afatinib, the radiation-enhanced

invasion was decreased, but the pretreatment with erlotinib did not show this effect.

Figure 6-1. Invasion assay of T24 and 5637 bladder cancer cells treated with irradiation +/-

afatinib (BIBW) or erlotinib (Tar)

Then we tested the expression of phosphorylated EGFR and HER2 as well as MMP-9

and MMP-2 in T24 and 5637 bladder cancer cells. As shown in Fig. 6-2, after irradiation the

expression of p-EGFR, p-HER2 and MMP-9 was increased, and the effect was decreased

after the pretreatment with afatinib. However the pretreatment with erlotinib did not show

this effect.

Figure 6-2. Western blot of T24 and 5637 bladder cancer cells treated with irradiation +/-

afatinib (BIBW) or erlotinib (Tarceva)

In addition, enzyme activity in culture media using zymography was also tested. The

result showed that after irradiation the activity of MMP-9 was increased, and the effect was

decreased after the pretreatment with afatinib. The influence of erlotinib pretreatment is

more variable in T24 and 5637 cells. In contrary, there was no obvious change in MMP-2

activity (Fig. 6-3).

Figure 6-3. Gelatin zymography of culture media from T24 and 5637 bladder cancer cells

treated with irradiation +/- afatinib (BIBW) or erlotinib (Tarceva)

Currently mechanistic and animal studies are ongoing to find out more ways to improve

the treatment outcome of bladder cancer radiotherapy.

相關文件