• Tidak ada hasil yang ditemukan

비영리 - S-Space - 서울대학교

N/A
N/A
Protected

Academic year: 2024

Membagikan "비영리 - S-Space - 서울대학교"

Copied!
41
0
0

Teks penuh

(1)

저작자표시-비영리-변경금지 2.0 대한민국 이용자는 아래의 조건을 따르는 경우에 한하여 자유롭게

l 이 저작물을 복제, 배포, 전송, 전시, 공연 및 방송할 수 있습니다. 다음과 같은 조건을 따라야 합니다:

l 귀하는, 이 저작물의 재이용이나 배포의 경우, 이 저작물에 적용된 이용허락조건 을 명확하게 나타내어야 합니다.

l 저작권자로부터 별도의 허가를 받으면 이러한 조건들은 적용되지 않습니다.

저작권법에 따른 이용자의 권리는 위의 내용에 의하여 영향을 받지 않습니다. 이것은 이용허락규약(Legal Code)을 이해하기 쉽게 요약한 것입니다.

Disclaimer

저작자표시. 귀하는 원저작자를 표시하여야 합니다.

비영리. 귀하는 이 저작물을 영리 목적으로 이용할 수 없습니다.

변경금지. 귀하는 이 저작물을 개작, 변형 또는 가공할 수 없습니다.

(2)

의학박사 학위논문

In vivo radiosensitizing and

immune modulatory effect of PI4K IIIα inhibition: role of PI3Kδ

PI4K IIIα 억제의 방사선 감작 효과 및 면역 조절 효과에 대한 in vivo 연구: PI3Kδ의 역할

2018 년 8 월

서울대학교 대학원 의학과 방사선종양학 전공

박 영 희

(3)

Abstract

In vivo radiosensitizing and

immune modulatory effect of PI4K IIIα inhibition: role of PI3Kδ

Younghee Park Department of Radiation Oncology, School of Medicine

The Graduate School Seoul National University

Background and Purpose: Phosphatidylinositol 4-phosphate (PI4P), produced by phosphatidylinositol 4-kinase (PI4K), is a common substrate for two signaling pathways involved in cell migration and proliferation; phospholipase C (PLC)/protein kinase C (PKC) pathway and phosphatidylinositol 3-kinase (PI3K)/Akt pathway. Overexpression of PI4Ks in various cancers have been reported and PI4K is suggested as a potential target for anticancer treatment.

Hepatitis C virus (HCV) uses the PI4K IIIα of host cells, which is one of the isozymes of PI4K, for its replication. One of the anti-HCV agents, 4-Anilino Quinazoline compound, exerts antiviral effect by inhibiting PI4K IIIα. In our recent report, we screened various anti-HCV agents and found that simeprevir had an inhibitory effect on PI4K IIIα in vitro. Simeprevir radiosensitized diverse human cancer cells by inhibiting both PLC/PKC and PI3K/Akt pathways. Recently, programmed death-1 (PD-1)/programmed death-ligand 1 (PD-L1) is identified as an important target for immunotherapy and the role of PI3K for regulation of PD-

(4)

L1 pathway has been suggested. Among the diverse PI3K isozymes, PI3Kδ is shown to have immune modulatory effect and has been investigated as a target for anticancer therapy in hematologic malignancies. Recent studies have reported the overexpression of PI3Kδ in solid tumors and antitumor effects of PI3Kδ inhibition.

Based on these previous studies, we investigated the radiosensitizing effect of simeprevir in in vivo tumor xenograft models and the mechanism of its interaction.

The immune modulatory effect of PI4K IIIα was evaluated in an immune- competent syngeneic murine tumor model.

Material and Methods: To investigate the radiosensitizing effect in vivo, breast and brain tumor xenograft models were constructed. BT474 cells were injected into the hind limb and U251 cells were injected into the caudate-putamen of BALB/c athymic nude mice. Mice were randomized into 4 groups; control, radiation (RT) alone, simeprevir alone and simeprevir plus RT. Simeprevir of 10 mg/kg was administered intraperitoneally three times a week for 2 weeks and radiation of 9 Gy in 3 fractions was delivered for 1 week. After completion of treatment, the tumor growth was measured at regular time interval. The tumor volume of breast tumor model was estimated using the formula (length × width × width)/2 and that of brain tumor model was measured with bioluminescence imaging. To evaluate the effect of PI4K IIIα inhibition and radiation on epithelial-mesenchymal transition (EMT) features, cell migration and invasion were evaluated in wound healing assays and modified Boyden chamber assays with BT474 and U251 cells.

Immunohistochemical staining (IHC) was performed in the tumor tissues harvested from the breast tumor xenograft model to evaluate the expression of PI3Kδ. The radiosensitizing effect of specific inhibition of PI3Kδ using RNA interference (RNAi) was evaluated in BT474 and MDA-MB-468 breast cancer cells using clonogenic assays. The effect of PI3Kδ inhibition by RNAi or simeprevir on PD- L1 expression was evaluated on Western blot analysis. To investigate the effect of simeprevir combined with PD-L1 inhibitor, clonogenic assays were performed with BT474 and MDA-MB-468 cells co-cultured with human T-lymphocytes. For

(5)

evaluation of immune modulatory effect of simeprevir in vivo, immune-competent syngeneic mouse model was established by injecting 4T1 murine breast cancer cells into the hind limb of immune-competent BALB/c mice. Mice were randomized into the 4 groups same as in the xenograft models. Delivered radiation dose was 8 Gy x 3 fractions in 1 week and simeprevir of 20mg/kg was administered intraperitoneally 3 times a week for 2 weeks. The tumor growth was measured with the same formula used in the breast xenograft model. Flow cytometry and Western blot assay were performed with harvested tissues.

Results: In a breast tumor xenograft model, PI4K IIIα inhibition using simeprevir combined with RT significantly delayed tumor growth compared to either treatment alone. In a brain tumor xenograft model, tumor volume was smaller in the group treated with simeprevir+RT than those in the other groups, but the difference was not statistically significant. PI4K IIIα inhibition by RNAi or simeprevir inhibited the cell migration and invasion on wound healing assay and modified Boyden chamber assay and the inhibitory effect was enhanced after combined treatment with radiation. On IHC, PI3Kδ expression was decreased in the group treated with simeprevir+RT. Specific inhibition of PI3Kδ using RNAi radiosensitized both BT474 and MDA-MB-468 cells and inhibited the PD-L1 and phosphorylated AKT (p-AKT) expressions. When various brain and breast tumor cell lines were screened, simeprevir down-regulated the PD-L1 expressions in U251 and MDA-MB-468 cells. PI4K IIIα inhibition by simeprevir enhanced the radiosensitizing effect of PD-L1 inhibitor in BT474 and MDA-MB-468 cells.

Combined treatment of simeprevir with PD-L1 inhibitor down-regulated the p- AKT. In an immune-competent syngeneic mouse model, the tumor growth was significantly delayed in the group treated with simeprevir+RT. When the tumor tissues from the immune-competent syngeneic mouse model were evaluated, the proportion of CD8(+) T cell was significantly increased and the expression of PI3Kδ was down-regulated after treatment with simeprevir+RT.

(6)

Conclusion: Inhibition of PI4K IIIα with simeprevir radiosensitized both in vivo tumor xenograft model and immune-competent syngeneic tumor model.

Simeprevir down-regulated the PI3Kδ expression and led to eversion of EMT features. PI4K IIIα inhibition had immune modulatory effect by regulating PD-L1 expression and affecting the CD4(+)/CD8(+) ratio of T lymphocyte through regulation of PI3Kδ. In conclusion, targeting PI4K IIIα with simeprevir is a viable drug repositioning approach for enhancing the therapeutic efficacy of radiation and effect of immune checkpoint blockades.

---

Keywords: Phosphatidylinositol 4-kinase IIIα, radiosensitivity, Phosphatidylinositol 3-kinase δ, immune checkpoint blockade

Student number: 2015-30597

(7)

Table of Contents

Abstract ... i

Table of Contents ... v

List of Figures ... vi

Introduction ... 1

Materials and Methods ... 3

Results ... 8

Discussion ... 21

References ... 25

Abstract in Korean ... 30

(8)

List of Figures

Figure 1. Radiosensitizing effect of simeprevir in an in vivo human breast tumor xenograft model ... 9 Figure 2. Radiosensitizing effect of simeprevir in an in vivo human brain

tumor xenograft model ... 10 Figure 3. Effect of PI4K IIIα inhibition on migration and invasion ... 12 Figure 4. Immunohistochemical staining of tissues harvested from in vivo

breast tumor xenograft model ... 14 Figure 5. Radiosensitizing effect of PI3Kδ inhibition using RNAi ... 14 Figure 6. Effect of PI3Kδ inhibition on expression of p-AKT and PD-L1 in

BT474 and MDA-MB-468 cells ... 16 Figure 7. Effect of simeprevir on PD-L1 expression in various brain and

breast tumor cell lines... ... 16 Figure 8. Effect of simeprevir on radiosensitizing effect of PD-L1 inhibitor

in BT474 and MDA-MB-468 cells ... 18 Figure 9. Radiosensitizing and immune modulatory effect of simeprevir in

an immune-competent syngeneic mouse model ... 20

(9)

Introduction

Phosphatidylinositol 4-phosphate (PI4P), produced by phosphatidylinositol 4- kinase (PI4K), is a common substrate for both phospholipase C (PLC)/protein kinase C (PKC) pathway and phosphatidylinositol 3-kinase (PI3K)/Akt pathway responsible for diverse cell functions and pathogenesis [1]. Four isotypes of PI4K, PI4K type II (α- and β-forms) and type III (α- and β-forms) have been identified and the involvement of PI4K IIIα in various cancers have been reported [2]. PI4K IIIα is associated with more invasive phenotypes in pancreatic cancer [3] and poor prognosis in hepatocellular carcinoma [4]. A recent study reported that upregulation of PI4K IIIα was associated with the migration and invasion of prostate cancer cells [5].

Recently, it has been reported that hepatitis C virus (HCV) uses PI4K IIIα of host cell for replication [6, 7]. Additionally, it is reported that the antiviral effect of 4-Anilino Quinazoline compound, one of the anti-HCV agent, is mediated through the inhibition of PI4K IIIα [8]. Based on previous studies, we screened various anti-HCV agents for the inhibitory effect on the PI4K IIIα activity and investigated the effect of PI4K IIIα inhibition on the cancer cells [9]. As a result, we identified that simeprevir, an anti-HCV agent, had an inhibitory effect on PI4K IIIα in various cancer cell lines and a significant radiosensitizing effect. Inhibition of PI4K IIIα by simeprevir resulted in down-regulation of p-PKC and p-AKT, suggesting that simeprevir is an effective anticancer agent that simultaneously inhibits two important pathways known to be involved in both tumorigenesis and treatment resistance. Moreover, simeprevir prolonged γH2AX foci and down-regulated phospho-DNA-PKcs after irradiation, suggesting that the radiosensitizing effects were mediated through impaired nonhomologous end-joining repair. These results suggested the successful drug repositioning of simeprevir as a new anticancer agent.

Based on these results, we investigated the radiosensitizing effect of PI4K IIIα inhibition in in vivo tumor models and evaluated the mechanisms of radiosensitization in this study.

(10)

Cancer immunotherapy has been widely investigated in various cancers [10].

Among the several target molecules, programmed death-1 (PD-1) and programmed death-ligand 1 (PD-L1) are important molecules currently under investigation and the radiosensitizing effects of the immune checkpoint inhibitors have been observed [11-13]. Although the mechanisms of immune checkpoint inhibitors are not fully understood, the PI3K/Akt pathway has been suggested to be closely involved in the PD-1/PD-L1 pathway [14-16].

Among the different isotypes of PI3K, PI3Kδ is mainly found in leukocytes and plays essential roles in the development and function of immune cells [17, 18].

The immune modulatory function of PI3Kδ has been reported and PI3Kδ have been investigated as a therapeutic target in hematologic malignancies [19-22].

Recent studies reported the high expression of PI3Kδ in various solid tumors and antitumor effect of PI3Kδ inhibition [23, 24]. In this study, we investigated the effect of PI4K IIIα inhibition on the expression of PI3Kδ and PD-L1 to evaluate the possible role of PI3Kδ inhibition as a strategy to enhance the effects of immunotherapy.

(11)

Materials and Methods Cell culture

The human breast cancer cell line (BT474, MDA-MB-231 and MDA-MB- 468), human malignant glioma cell line (T98G, U87, U251) and murine breast cancer cell line (4T1) purchased from the American Type Culture Collection (Manassas, VA, USA) were cultured at 37°C in 5% CO2 humidified chambers.

Cells were maintained in Dulbecco Modified Eagle Medium (Life Technology, Inc., Carlsbad, CA, USA) or Roswell Park Memorial Institute 1640 (Life Technology, Inc.) supplemented with 10% fetal bovine serum. When evaluating the immune modulatory effect in vitro, the tumor cells were co-cultured with activated Jurkat T cells.

In vivo tumor xenograft model

Breast tumor model

By injecting 5 × 106 BT474 cells into the hind limb of 6- to 8-week-old BALB/c athymic nude mice (Orient Bio, Inc., Sungnam, Korea), human breast cancer xenografts were established. On day 25 after injection, when the longest diameter of tumor exceeds 10mm, mice were randomized into 4 groups, control, simeprevir, radiation (RT), and simeprevir plus RT with 4 mice per group.

Simeprevir (10 mg/kg) was administered intraperitoneally three times weekly for 2 weeks. During the second week of drug treatment, mice were irradiated three times with 3 Gy for a total of 9 Gy using a 6 MeV electron beam with a 1 cm bolus applied 3 h after simeprevir injection. Lead blocks were used to shield the non- tumor part of the mice. After treatment was complete, tumor size was measured weekly by two investigators for 2 months after grouping. The tumor volume was estimated using the formula (length × width × width)/2. All procedures involving mice were performed according to a protocol approved by the Institutional Animal Care and Use Committee at the Clinical Research Institute, Seoul National

(12)

University Bundang Hospital.

Brain tumor model

U251 cells were transfected with a pGL4.5 luciferase reporter vector (Promega, Madison, WI, USA) according to the manufacturer’s protocol. BALB/c athymic nude mice were anesthetized and U251 cells (3 × 105 cells) were injected into the caudate-putamen with a 26-G needle attached to a Hamilton syringe. Two weeks after injection, 12 mice were randomized into 4 groups: control, simeprevir, RT, and simeprevir plus RT. Simeprevir was administered with the same dose and schedule as in the breast tumor xenograft model. The radiation of 9 Gy in 3 fractions was delivered in first week of treatment before simeprevir injection to increase the permeability of blood brain barrier (BBB). Tumor burden was measured with bioluminescence imaging. Because of the dead mice, the growth curves of each groups could not be obtained. Instead, the relative tumor volumes measured when all the mice were alive (on day 39 after grouping) were compared.

Syngeneic mouse model

The 4T1 murine triple negative breast cancer cells (6 × 105 cells) were injected into the hind limb of BALB/c immune-competent mice (Orient Bio, Inc).

After 12 days after injection, when the longest diameter of tumor exceeds 10mm, 16 mice were randomized into 4 groups; control, simeprevir, radiation (RT), and simeprevir plus RT. Simeprevir (20mg/kg) and radiation (8 Gy × 3 fractions) were delivered in the same way as described in breast tumor xenograft model. Mice were sacrificed at 1 month after tumor inoculation and tumors were harvested.

Bioluminescence imaging (BLI)

Bioluminescence imaging was performed using the IVIS Imaging System 100 series (Xenogen Corporation, Alameda, CA, USA) according to the manufacturer’s

(13)

protocol. Mice were injected with 150 mg/kg D-luciferin intraperitoneally and after 15 min, anesthetized with 1–2% isoflurane. Images were acquired between 5 and 10 min and peak luminescence signals were recorded. To compensate initial small differences in tumor burdens, the relative tumor burdens were defined as (signal value at the time of last measurement – signal value at baseline)/signal value at baseline; these values were used for statistical analysis.

Immunohistochemical (IHC) staining

The tissues harvested from mice were fixed with formalin and paraffin- embedded tissue blocks were constructed. Tissue sections were deparaffinized and rehydrated according to standard procedures. Staining was conducted using primary antibody against PI3Kδ (Abcam, Cambridge, UK).

Wound healing assay

Cells were grown to confluence in 6-well plates (Sonic-Seal Slide, Nalgene Nunc, Rochester, NY, USA) and then starved by culturing in DMEM without fetal bovine serum for 24 h. Each well was then divided into a 2 × 3 grid. Using a 1-mL pipette tip, a line was scratched in each hemisphere of the well to wound the cells, and the medium was changed to starvation medium. Images were taken of the intersections of linear cell wounds and grid lines. Images were captured immediately after wounding and after 24 h. The distance between wound edges was measured using Image J software (NIH, Bethesda, MD, USA).

Modified Boyden chamber assay

A Transwell system (Corning, Rochester, NY, USA), which allows cells to migrate through 8 μm pores in polycarbonate membranes was used to evaluate the cell invasion. After trypsinization, an aliquot of 104 cells was added to the upper chamber. After 24 hours, inserts were fixed in methanol and stained with 1%

(14)

crystal violet. The invading cells on the lower surface were counted.

RNA interference (RNAi)

Cells were plated in 6-well culture plates and transfected with PI3Kδ-specific (GE Dharmacon, Lafayette, CO, USA) or nonspecific RNA using Lipofectamine RNAiMAX transfection reagent (Invitrogen, Carlsbad, CA, USA) in reduced- serum medium (OPTIMEM, Life Technologies) according to the manufacturer’s protocol.

Clonogenic assay

Identical numbers of cells were plated into 6-well culture plate across the different treatment groups for each radiation dose. Forty-eight hours after treatment, the cells were irradiated with 6 megavoltage X-ray from a linear accelerator (Varian Medical System, Palo Alto, CA, USA) at a dose rate of 2.46 Gy/min and then incubated for colony formation for 14–23 days. Colonies were fixed with methanol and stained with 0.5% crystal violet. The colonies containing 50 or more cells were counted and the surviving fraction was calculated. The radiation-survival data was fitted to a linear-quadratic model using Kaleidagraph version 3.51 (Synergy Software, Reading, PA, USA). A sensitizer enhancement ratio of 0.05 (SER0.05) was calculated as the ratio of the dose resulting in a surviving fraction of 0.05 without drugs to that with drugs.

Western blot analysis

Cells were washed, scraped, and resuspended in lysis buffer (Cell Signaling Technology, Danvers, MA, USA). Proteins were solubilized by sonication, separated by SDS-PAGE, and electroblotted onto polyvinylidenedifluoride membranes (Millipore). Membranes were blocked in Tris-buffered saline and Tween-20 solution containing 5% skim milk or bovine serum albumin and then

(15)

probed with a primary antibody directed against PI3Kδ (Abcam), p-AKT (Cell Signaling Technology), PD-L1 (Biolegend, San Diego, CA, USA), and β-actin (Santa Cruz Biotechnology, Santa Cruz, CA, USA). After washing and blocking, membranes were incubated with peroxidase-conjugated goat anti-rabbit or anti- mouse IgG secondary antibody (Jackson ImmunoResearch Laboratories, West Grove, PA, USA) at dilutions of 1:10,000 for 1 h.

Flow cytometry analysis

Tumor cells were dissociated into single cell suspension and stained with CD45, CD4 and CD8 antibodies. Cells were analyzed and sorted using a FACS Calibur flow cytometer (BD Biosciences) and FACS Aria sorters (BD Biosciences).

Statistical Analysis

Statistical analysis was performed with SPSS version 18.0 (SPSS, Inc., Chicago, IL, USA). The t-test and one way analysis of variance were used to compare the mean values between groups.

(16)

Results

Part 1. Radiosensitizing effect of PI4K IIIα inhibition

1-1. In vivo tumor growth delay with PI4K IIIα inhibition and radiation

In our recent study, the radiosensitizing effect of simeprevir was demonstrated in various human cancer cell lines. To investigate whether radiosensitization by simeprevir occurs in vivo, we constructed in vivo tumor xenograft models of BT474 breast cancer and U251 brain tumor cells, and the effects of simeprevir on tumor growth were investigated.

Tumor volume curves for the control and groups treated with simeprevir, RT, or simeprevir plus RT in breast cancer model are shown in Figure 1. Tumor growth was significantly delayed in the simeprevir plus RT group compared with the other three groups (p<0.05). Differences in tumor growth were evident beginning at 13 days after grouping and persisted until the final measurement.

The effect of simeprevir on the brain tumor model is presented in Figure 2. In the simeprevir plus RT group, bioluminescence imaging (BLI) showed the greatest decrease in signals and the relative tumor burden was lowest, although the differences were not statistically significant. According to our previous results, the radiosensitizing effect of simeprevir was evident in vitro in the U251 brain tumor cell line, but this was not observed in the in vivo model.

(17)

Figure 1. Radiosensitizing effect of simeprevir in an in vivo human breast tumor xenograft model. Tumor volume curves of each group; control, simeprevir alone, radiation alone, and simeprevir+radiation. Tumor growth was significantly delayed in the group treated with simeprevir plus radiation in BT474 breast cancer model (P<0.05). No significant differences between control, simeprevir alone, and radiation alone were found. RT = radiation

(18)

Figure 2. Radiosensitizing effect of simeprevir in an in vivo human brain tumor xenograft model. (A) Representative bioluminescence images of human brain tumor xenografts when all 3 mice were alive (On day 39 after grouping). (B) The relative tumor burden was lower in the group treated with simeprevir and radiation compared to in the other groups treated with either treatment alone, but the differences were not statistically significant. Bar represents standard error. Sim

= simeprevir, RT = radiation

(19)

1-2. Effect of PI4K IIIα inhibition on epithelial-mesenchymal transition

To investigate the effect of PI4K IIIα inhibition by simeprevir on the epithelial-mesenchymal transition (EMT), migration and invasion of U251 and BT474 cells were evaluated. In a wound healing assay to evaluate the cell migration, pretreatment with PI4K IIIα small interfering RNA (siRNA) or simeprevir markedly inhibited the migration potential of both cancer cells (Figure 3-A). The inhibitory effects of PI4K IIIα inhibition on migration potential were enhanced when combined with radiation. Similarly, the invasion rate of both cancer cell lines were significantly compromised after treatment with PI4K IIIα siRNA or simeprevir and the effects were more prominent when combined with radiation (Figure 3-B).
(20)

Figure 3. Effect of PI4K IIIα inhibition on migration and invasion. Migration and invasion potential of U251 cells and BT474 cells were assessed with wound healing assays (A) and modified Boyden chamber assay (B), respectively. Stained cells were analyzed in representative fields (x100). Selective inhibition of PI4K IIIα using siRNA inhibited cell migration and invasion in both cell lines and similar results were found after treatment with 200 nM simeprevir. The inhibition of cell migration and invasion was more prominent after combined treatment of PI4K IIIα inhibition and radiation. Bar and asterisk represent standard error and statistical significance of p<0.05 compared with control, respectively. Sim = simeprevir, RT = radiation

(21)

1-3. Radiosensitizing effect of PI4K IIIα inhibition was mediated by PI3Kδ inhibition

After confirmation of the radiosensitizing effect of simeprevir, we performed immunohistochemical staining using cancer tissues harvested from breast xenograft model to evaluate the effect of PI4K IIIα inhibition on the PI3Kδ isotype. We found that PI3Kδ expression was decreased in tumors treated with simeprevir plus RT compared to tumors from the other groups (Figure 4). This suggests that the radiosensitizing effect of PI4K IIIα inhibition is mediated by down-regulation of PI3Kδ.

Next, to investigate whether inhibition of PI3Kδ increases radiosensitivity of cancer cells, we performed clonogenic assays using RNAi for PI3Kδ. As shown in Figure 5, specific inhibition of PI3Kδ using siRNA significantly enhanced radiation-induced cell killing in BT474 (SER0.05 1.16) and MDA-MB-468 (SER0.05 1.28) breast cancer cells.

(22)

Figure 4. Immunohistochemical staining of tissues harvested from in vivo breast tumor xenograft model. Expression of PI3Kδ was down-regulated in tumors treated with simeprevir plus radiation (x200). RT = radiation

Figure 5. Radiosensitizing effect of PI3Kδ inhibition using RNAi. Specific inhibition of PI3Kδ using RNA interference (RNAi) increased radiosensitivity of BT474 and MDA-MB-468 cells.

(23)

Part 2. Immune modulatory effect of PI4K IIIα inhibition

2-1. PI4K IIIα inhibition down-regulated PD-L1 expression thorough modulation of PI3Kδ

It has been reported that PI3Kδ has immune modulatory functions associated with PD-L1 expression. Therefore, we evaluated whether PI3Kδ inhibition could modulate PD-L1 expressions in BT474 and MDA-MB-468 breast cancer cells.

Specific PI3Kδ inhibition using siRNA led to significant down-regulation of PD- L1 expression in MDA-MB-468 cells although the effect of PI3Kδ inhibition on PD-L1 expression was not prominent in BT474 cells. In both cell lines, PI3Kδ inhibition down-regulated the expression of p-AKT (Figure 6).

To determine whether PI3Kδ inhibition through PI4K IIIα inhibition affects PD-L1 expression, the effect of simeprevir on PD-L1 expression was evaluated in various cancer cell lines including T98G, U87, and U251 glioma cell lines and BT474, MDA-MB-231, and MDA-MB-468 breast cancer cell lines (Figure 7). In U251 and MDA-MB-468 cell lines, simeprevir significantly inhibited PD-L1 expression.

These results suggest that PI4K IIIα inhibition down-regulates PD-L1 expression by modulating PI3Kδ and p-AKT.

(24)

Figure 6. Effect of PI3Kδ inhibition on expression of p-AKT and PD-L1 in BT474 and MDA-MB-468 cells. PI3Kδ inhibition using RNAi significantly decreased p-AKT expression in both cell lines and PD-L1 expression in MDA-MB- 468 cells.

Figure 7. Effect of simeprevir on PD-L1 expression in various brain and breast tumor cell lines. In U251 and MDA-MB-468 cells, simeprevir (500 nM) significantly inhibited expression of PD-L1.

(25)

2-2. Combination of simeprevir and PD-L1 inhibitor enhanced radiation-induced cell death through modulation of PI3Kδ

After the effects of PI4K IIIα on PD-L1 expressions were determined, we performed clonogenic assays to assess the effect of PI4K IIIα on the radiosensitizing effect of immune checkpoint blockades in BT474 and MDA-MB- 468 cells (Figure 8). Simeprevir combined with PD-L1 inhibitor significantly enhanced radiation-induced cell killing (SER0.05 1.22 in BT 474 cells, 1.34 in MDA-MB-468 cells) compared to either treatment alone (SER0.05 for simeprevir and PD-L1 inhibitor, 1.13 and 1.08 in BT474 cells, 1.11 and 1.26 in MDA-MB-468 cells, respectively). PD-L1 inhibitor or simprevir alone inhibited p-AKT expression in both cell lines as is shown in the results for PI3Kδ siRNA experiment and a combination of the two agents led to the strongest down-regulation of p-AKT.

(26)

Figure 8. Effect of simeprevir on radiosensitizing effect of PD-L1 inhibitor in BT474 and MDA-MB-468 cells. Simeprevir and PD-L1 inhibitor alone increased radiation-induced cell death and down-regulated the expression of p-AKT.

Combined treatment with the two agents showed enhanced radiosensitizing effects and down-regulation of p-AKT expression.

(27)

2-3. Radiosensitizing and immune modulatory effect of PI4K IIIα inhibition in immune-competent syngeneic mouse model

To evaluate the immune modulatory effect of simeprevir, syngeneic tumor model using immune-competent mice were constructed. The treatment groups were same as in the experiment of xenograft models; control, simeprevir, RT, or simeprevir plus RT. Simeprevir showed significant radiosensitizing effect in the syngeneic murine tumor model (Figure 9-A). To investigate the immune modulatory effect, flow cytometric analyses were performed using the tumor harvested from syngeneic mouse model. As a result (Figure 9-B), the proportion of CD8(+) T cell was significantly increased after treatment with simeprevir combined with RT. PI3Kδ expression was down-regulated after treatment with simeprevir combined with RT (Figure 9-C), suggesting that PI4K IIIα inhibition exhibits immune-modulation via down-regulation of PI3Kδ expression.

(28)

Figure 9. Radiosensitizing and immune modulatory effect of simeprevir in an immune-competent syngeneic mouse model. Tumor volume curves showed significant tumor growth delay in the group treated with simeprevir and radiation in immune-competent syngeneic mouse model (P < 0.05). Relative tumor volume = tumor volume divided at specific time by tumor volume at randomization. Tumor cells harvested from mice were analyzed in fluorescence-activated cell sorting (B) and western blotting (C). After treatment with simeprevir and radiation, the proportion CD8(+) T cell was significantly increased and the expression of PI3Kδ was down-regulated. Sim = simeprevir, RT = radiation

(29)

Discussion

Our previous in vitro study [9] reported the radiosensitizing effect of PI4K IIIα inhibition. Simeprevir, an anti-HCV agent, inhibited PI4K IIIα activity and increased radiation-induced cell killing in a panel of human cancer cell lines.

Pretreatment with simeprevir led to prolongation of radiation-induced γH2AX foci and down-regulation of phospho-DNA-PKcs, indicating impaired DNA double- strand break repair. Apoptosis and autophagy were the major modes of cell death.

Based on these results, we confirmed the radiosensitizing effect of PI4K IIIα inhibition in in vivo tumor xenograft models in this study. Combined treatment with simeprevir and radiation significantly attenuated tumor growth in breast cancer xenograft model, while tumor growth in the radiation or simeprevir alone groups was similar to in the control group. This suggests that the dose of radiation delivered in our study, 9 Gy in 3 fractions, was not sufficient to delay tumor growth; however, when combined with simeprevir, the therapeutic efficacy of radiation was dramatically enhanced. A high dose of radiation increases tumor control, but toxicity to normal tissue also increases. Radiosensitizers were previously suggested to overcome this limitation, but the toxicity of radiosensitizers limited their widespread use. Simeprevir is an anti-HCV agent that has been shown to be safe in humans and is widely used to treat HCV without significant toxicity. Therefore, PI4K IIIα inhibition using simeprevir may be a viable and safe strategy for enhancing the cytotoxic effect of radiation therapy.

Further studies comparing the efficacy of simeprevir with the other chemotherapeutic agents and identifying the subset of patients who could benefit from simeprevir would be needed.

In the brain tumor xenograft model, the tendency for tumor growth delay with simeprevir plus RT was observed, but the results were not statistically significant.

In an in vitro experiment, simeprevir showed a significant radiosensitizing effect in U251 brain tumor cells [9]. Simeprevir was initially developed as an agent targeting HCV; the molecular weight is 749.9 Da (g/mol) and the BBB

(30)

penetrability of simeprevir was not investigated. As is well-known, the BBB impairs effective delivery of a wide range of anticancer agents to the brain up to the therapeutic level. The less discernible radiosensitizing effect in the in vivo brain tumor model in this study may be explained by the limited penetration of simeprevir into the tumor in the brain because of its molecular weight. Although we delivered radiation before administration of simeprevir in the brain tumor model to increase the permeability of the BBB, a significant radiosensitizing effect was not observed. Further studies using an anti-PI4K IIIα agent that can cross the BBB up to a therapeutic level would confirm the radiosensitizing effect of PI4K IIIα inhibition in brain tumor xenograft model.

The radiosensitizing effects of PI3K inhibition have been widely reported [25- 29]. Recently, a specific inhibitor of PI3Kδ, IC486068, was shown to enhance the radiosensitivity of tumor cells [30]. PI3Kδ, one of the isotypes in the class I PI3K family, which is expressed mainly in lymphoid cells, has been reported to be highly expressed in solid tumors as well as hematologic malignancies [23, 24].

Additionally, it was reported that overexpression of PI3Kδ resulted in constitutive Akt activation and induced oncogenic transformation of normal cells [31]. In our study, PI4K IIIα inhibition combined with radiation using simeprevir down- regulated PI3Kδ expression. Furthermore, specific inhibition of PI3Kδ using siRNA enhanced radiation-induced cell killing, suggested that the mechanism of radiosensitization is related to PI3Kδ inhibition. To our knowledge, this is the second study reporting the radiosensitizing effect of PI3Kδ inhibition; the first study was reported by Geng et al [30]. However, in their study, the suggested mechanism of radiosensitization was enhanced radiation-induced apoptosis of endothelial cells and impaired tumor vascular formation by PI3Kδ inhibition, but not a direct cytotoxic effect to cancer cells. Therefore, this is the first study reporting enhanced radiation-induced tumor cell killing by PI3Kδ inhibition, and PI3Kδ may be a potential target for radiosensitization by simultaneously regulating the radiation effect on tumor cells and tumor microenvironment.

(31)

The tumor microenvironment is known to be important in tumorigenesis and responsible for treatment resistance [32, 33]. EMT is an essential process for cancer cells to acquire invasiveness and metastatic potential and has been suggested as a promising target for modulating the tumor microenvironment [34, 35]. In our study, we found that PI4KIIIα inhibition led to eversion of EMT as indicated by impaired cell migration and invasion. Geng et al. showed that that PI3Kδ inhibited the tumor vasculature by enhancing radiation-induced cell death of endothelial cells and inhibited endothelial cell migration [30]. Another study by Sawyer et al. demonstrated that PI3Kδ is associated with breast cancer cell migration, regulating both directionality and migration speed, and confers selective migratory capacities (chemotaxis) of breast cancer cells [23]. Therefore, we predict that PI4K IIIα inhibition controls the eversion of EMT via PI3Kδ inhibition.

Immunotherapy is an emerging anticancer treatment, and enhanced cancer cell killing by radiation when combined with immune checkpoint blockades have been reported [13, 36]. Although the specific mechanism of immune checkpoint blockades are not fully understood, some studies suggested that the PI3K pathway is involved in PD-L1 expression and immune-resistance in tumor cells [15, 37, 38].

Previously, Ali et al. showed that PI3Kδ inactivation inhibited tumor growth in an immune-competent syngeneic murine tumor model through enhanced cytotoxic T cell functions by disrupting regulatory T cell function, which is known to be essential for immune evasion of tumors [39]. It also has been demonstrated that PI3Kδ is required for cytotoxic T cell functions involving the proliferation of cytotoxic T cells and degranulation process [40]. Kan-o et al. reported that PI3Kδ up-regulated PD-L1 in airway epithelial cells, which is an immune evasion strategy used by respiratory viruses [41]. Therefore, we further investigated the mechanism of radiosensitization of PI4K IIIα inhibition, focusing on immune modulation through PI3Kδ.

As a result, specific inhibition of PI3Kδ using RNAi decreased PD-L1 expression and AKT expression. Simeprevir showed inhibitory effect on PD-L1

(32)

expression in MDA-MB-468 and U251 cells. Furthermore, PI4K IIIα inhibition enhanced the radiosensitizing effect of PD-L1 blockade. This is the first study to demonstrate the association of PI3Kδ with PD-L1 in tumor cells. Therefore, the immune modulatory effect of simeprevir is suggested and the modulation of PI3Kδ appears to be the underlying mechanism.

Although we identified immune modulatory effect of PI4K IIIα inhibition in in vitro human cancer cells co-cultured with human T lymphocytes, the findings provided only limited mechanistic interpretation. To overcome this issue, we performed in vivo study using immune-competent syngeneic murine tumor model.

PI4K IIIα inhibition using simeprevir showed significant radiosensitizing effect in syngeneic tumor model. When evaluating the tumor cells harvested from murine tumor sample, the proportion of CD8(+) T lymphocyte was significantly increased while that of CD4(+) T lymphocyte was not significantly changed. Many studies have reported the association of tumor infiltrating CD8(+) lymphocytes with prognosis [39, 40] and prognostic value of CD4(+)/CD8(+) ratio of tumor infiltrating lymphocytes [41, 42]. We can speculate that simeprevir can increase anticancer immune response by affecting the CD4(+)/CD8(+) ratio of T lymphocyte.

In conclusion, we confirmed the radiosensitizing effect of PI4K IIIα inhibition using simeprevir in in vivo tumor xenograft model and immune-competent syngeneic mouse model. Simeprevir radiosensitized tumor cells by modulating PI3Kδ and led to eversion of EMT features. PI4K IIIα inhibition modulated the immune response by regulating PD-L1 expression and affecting the CD4(+)/CD8(+) ratio of T lymphocyte. Taken together, targeting of PI4K IIIα may enhance the cytotoxic effect of radiation by modulating anti-tumor immunity and the tumor microenvironment.

(33)

Reference

[1] E. L. Clayton, S. Minogue and M. G. Waugh. Mammalian phosphatidylinositol 4-kinases as modulators of membrane trafficking and lipid signaling networks. Prog Lipid Res 2013;52(3):294-304.

[2] M. G. Waugh. Phosphatidylinositol 4-kinases, phosphatidylinositol 4- phosphate and cancer. Cancer Lett 2012;325(2):125-31.

[3] S. Ishikawa, H. Egami, T. Kurizaki, J. Akagi, Y. Tamori, N. Yoshida, et al.

Identification of genes related to invasion and metastasis in pancreatic cancer by cDNA representational difference analysis. J Exp Clin Cancer Res 2003;22(2):299- 306.

[4] A. Ilboudo, J. C. Nault, H. Dubois-Pot-Schneider, A. Corlu, J. Zucman- Rossi, M. Samson, et al. Overexpression of phosphatidylinositol 4-kinase type IIIalpha is associated with undifferentiated status and poor prognosis of human hepatocellular carcinoma. BMC Cancer 2014;14:7.

[5] D. Sbrissa, L. Semaan, L. Yanfeng, A. Shisheva and S. R. Chinni.

Abstract 5162: Phosphatidylinositol 4-Kinase type IIIa (PI4KA) expression in prostate cancer. Cancer Research 2015;75(15 Supplement):5162-.

[6] K. L. Berger, J. D. Cooper, N. S. Heaton, R. Yoon, T. E. Oakland, T. X.

Jordan, et al. Roles for endocytic trafficking and phosphatidylinositol 4-kinase III alpha in hepatitis C virus replication. Proc Natl Acad Sci U S A 2009;106(18):7577-82.

[7] J. Borawski, P. Troke, X. Puyang, V. Gibaja, S. Zhao, C. Mickanin, et al.

Class III phosphatidylinositol 4-kinase alpha and beta are novel host factor regulators of hepatitis C virus replication. J Virol 2009;83(19):10058-74.

[8] A. Bianco, V. Reghellin, L. Donnici, S. Fenu, R. Alvarez, C. Baruffa, et al.

Metabolism of phosphatidylinositol 4-kinase IIIalpha-dependent PI4P Is subverted by HCV and is targeted by a 4-anilino quinazoline with antiviral activity. PLoS Pathog 2012;8(3):e1002576.

[9] J. Kwon, D. H. Kim, J. M. Park, Y. H. Park, Y. H. Hwang, H. G. Wu, et al.

(34)

Targeting Phosphatidylinositol 4-Kinase IIIalpha for Radiosensitization: A Potential Model of Drug Repositioning Using an Anti-Hepatitis C Viral Agent. Int J Radiat Oncol Biol Phys 2016;96(4):867-76.

[10] K. M. Mahoney, P. D. Rennert and G. J. Freeman. Combination cancer immunotherapy and new immunomodulatory targets. Nat Rev Drug Discov 2015;14(8):561-84.

[11] J. Chen, C. C. Jiang, L. Jin and X. D. Zhang. Regulation of PD-L1: a novel role of pro-survival signalling in cancer. Ann Oncol 2016;27(3):409-16.

[12] A. B. Sharabi, M. Lim, T. L. DeWeese and C. G. Drake. Radiation and checkpoint blockade immunotherapy: radiosensitisation and potential mechanisms of synergy. Lancet Oncol 2015;16(13):e498-509.

[13] L. Deng, H. Liang, B. Burnette, M. Beckett, T. Darga, R. R.

Weichselbaum, et al. Irradiation and anti-PD-L1 treatment synergistically promote antitumor immunity in mice. J Clin Invest 2014;124(2):687-95.

[14] A. T. Parsa, J. S. Waldron, A. Panner, C. A. Crane, I. F. Parney, J. J.

Barry, et al. Loss of tumor suppressor PTEN function increases B7-H1 expression and immunoresistance in glioma. Nat Med 2007;13(1):84-8.

[15] M. Atefi, E. Avramis, A. Lassen, D. J. Wong, L. Robert, D. Foulad, et al.

Effects of MAPK and PI3K pathways on PD-L1 expression in melanoma. Clin Cancer Res 2014;20(13):3446-57.

[16] M. Song, D. Chen, B. Lu, C. Wang, J. Zhang, L. Huang, et al. PTEN loss increases PD-L1 protein expression and affects the correlation between PD-L1 expression and clinical parameters in colorectal cancer. PLoS One 2013;8(6):e65821.

[17] W. P. Fung-Leung. Phosphoinositide 3-kinase delta (PI3Kdelta) in leukocyte signaling and function. Cell Signal 2011;23(4):603-8.

[18] D. Chantry, A. Vojtek, A. Kashishian, D. A. Holtzman, C. Wood, P. W.

Gray, et al. p110delta, a novel phosphatidylinositol 3-kinase catalytic subunit that associates with p85 and is expressed predominantly in leukocytes. J Biol Chem

(35)

1997;272(31):19236-41.

[19] J. A. Burger and K. Okkenhaug. Haematological cancer: idelalisib- targeting PI3Kdelta in patients with B-cell malignancies. Nat Rev Clin Oncol 2014;11(4):184-6.

[20] A. K. Gopal, B. S. Kahl, S. de Vos, N. D. Wagner-Johnston, S. J. Schuster, W. Jurczak, et al. Mature Follow up from a Phase 2 Study of PI3K-Delta Inhibitor Idelalisib in Patients with Double (Rituximab and Alkylating agent)-Refractory Indolent B-Cell Non-Hodgkin Lymphoma (iNHL). Blood 2014;124(21):1708-.

[21] O. A. O'Connor, I. W. Flinn, M. R. Patel, T. S. Fenske, C. Deng, D. M.

Brander, et al. TGR-1202, a Novel Once Daily PI3K-Delta Inhibitor, Demonstrates Clinical Activity with a Favorable Safety Profile in Patients with CLL and B-Cell Lymphoma. Blood 2015;126(23):4154-.

[22] Q. Yang, P. Modi, T. Newcomb, C. Queva and V. Gandhi. Idelalisib:

First-in-Class PI3K Delta Inhibitor for the Treatment of Chronic Lymphocytic Leukemia, Small Lymphocytic Leukemia, and Follicular Lymphoma. Clin Cancer Res 2015;21(7):1537-42.

[23] C. Sawyer, J. Sturge, D. C. Bennett, M. J. O'Hare, W. E. Allen, J. Bain, et al. Regulation of breast cancer cell chemotaxis by the phosphoinositide 3-kinase p110delta. Cancer Res 2003;63(7):1667-75.

[24] N. Tzenaki, M. Andreou, K. Stratigi, A. Vergetaki, A. Makrigiannakis, B.

Vanhaesebroeck, et al. High levels of p110delta PI3K expression in solid tumor cells suppress PTEN activity, generating cellular sensitivity to p110delta inhibitors through PTEN activation. Faseb j 2012;26(6):2498-508.

[25] E. J. Choi, B. J. Cho, D. J. Lee, Y. H. Hwang, S. H. Chun, H. H. Kim, et al. Enhanced cytotoxic effect of radiation and temozolomide in malignant glioma cells: targeting PI3K-AKT-mTOR signaling, HSP90 and histone deacetylases.

BMC Cancer 2014;14:17.

[26] M. No, E. J. Choi and I. A. Kim. Targeting HER2 signaling pathway for radiosensitization: alternative strategy for therapeutic resistance. Cancer Biol Ther

(36)

2009;8(24):2351-61.

[27] N. Y. Jang, D. H. Kim, B. J. Cho, E. J. Choi, J. S. Lee, H. G. Wu, et al.

Radiosensitization with combined use of olaparib and PI-103 in triple-negative breast cancer. BMC Cancer 2015;15:89.

[28] M. Toulany, M. Iida, S. Keinath, F. F. Iyi, K. Mueck, B. Fehrenbacher, et al. Dual targeting of PI3K and MEK enhances the radiation response of K-RAS mutated non-small cell lung cancer. Oncotarget 2016;7(28):43746-61.

[29] M. Toulany and H. P. Rodemann. Phosphatidylinositol 3-kinase/Akt signaling as a key mediator of tumor cell responsiveness to radiation. Semin Cancer Biol 2015;35:180-90.

[30] L. Geng, J. Tan, E. Himmelfarb, A. Schueneman, K. Niermann, J. Brousal, et al. A specific antagonist of the p110delta catalytic component of phosphatidylinositol 3'-kinase, IC486068, enhances radiation-induced tumor vascular destruction. Cancer Res 2004;64(14):4893-9.

[31] S. Kang, A. Denley, B. Vanhaesebroeck and P. K. Vogt. Oncogenic transformation induced by the p110beta, -gamma, and -delta isoforms of class I phosphoinositide 3-kinase. Proc Natl Acad Sci U S A 2006;103(5):1289-94.

[32] E. L. Heinrich, T. C. Walser, K. Krysan, E. L. Liclican, J. L. Grant, N. L.

Rodriguez, et al. The inflammatory tumor microenvironment, epithelial mesenchymal transition and lung carcinogenesis. Cancer Microenviron 2012;5(1):5-18.

[33] D. Gao, L. T. Vahdat, S. Wong, J. C. Chang and V. Mittal.

Microenvironmental regulation of epithelial-mesenchymal transitions in cancer.

Cancer Res 2012;72(19):4883-9.

[34] C. J. Creighton, D. L. Gibbons and J. M. Kurie. The role of epithelial- mesenchymal transition programming in invasion and metastasis: a clinical perspective. Cancer Manag Res 2013;5:187-95.

[35] H. Y. Jung, L. Fattet and J. Yang. Molecular pathways: linking tumor microenvironment to epithelial-mesenchymal transition in metastasis. Clin Cancer

(37)

Res 2015;21(5):962-8.

[36] S. J. Dovedi, A. L. Adlard, G. Lipowska-Bhalla, C. McKenna, S. Jones, E.

J. Cheadle, et al. Acquired resistance to fractionated radiotherapy can be overcome by concurrent PD-L1 blockade. Cancer Res 2014;74(19):5458-68.

[37] C. A. Crane, A. Panner, J. C. Murray, S. P. Wilson, H. Xu, L. Chen, et al.

PI(3) kinase is associated with a mechanism of immunoresistance in breast and prostate cancer. Oncogene 2009;28(2):306-12.

[38] X. Jiang, J. Zhou, A. Giobbie-Hurder, J. Wargo and F. S. Hodi. The activation of MAPK in melanoma cells resistant to BRAF inhibition promotes PD- L1 expression that is reversible by MEK and PI3K inhibition. Clin Cancer Res 2013;19(3):598-609.

[39] S. Hadrup, M. Donia and P. Thor Straten. Effector CD4 and CD8 T cells and their role in the tumor microenvironment. Cancer Microenviron 2013;6(2):123-33.

[40] S. M. Mahmoud, E. C. Paish, D. G. Powe, R. D. Macmillan, M. J.

Grainge, A. H. Lee, et al. Tumor-infiltrating CD8+ lymphocytes predict clinical outcome in breast cancer. J Clin Oncol 2011;29(15):1949-55.

[41] A. C. Diederichsen, J. Hjelmborg, P. B. Christensen, J. Zeuthen and C.

Fenger. Prognostic value of the CD4+/CD8+ ratio of tumour infiltrating lymphocytes in colorectal cancer and HLA-DR expression on tumour cells. Cancer Immunol Immunother 2003;52(7):423-8.

[42] W. Shah, X. Yan, L. Jing, Y. Zhou, H. Chen and Y. Wang. A reversed CD4/CD8 ratio of tumor-infiltrating lymphocytes and a high percentage of CD4(+)FOXP3(+) regulatory T cells are significantly associated with clinical outcome in squamous cell carcinoma of the cervix. Cell Mol Immunol 2011;8(1):59-66.

(38)

국문초록

배경 및 목적: 포스파티딜이노시톨 4-키나아제 (PI4K)에 의하여 형성되는 포스파티딜이노시톨 4-포스페이트 (PI4P)는 세포의 이동과 증식에 관련된 두 가지 신호경로인 phospholipase C (PLC)/protein kinase C (PKC) 경로와 phosphatidylinositol 3-kinase (PI3K)/Akt 경로에 공통적인 기질이다. 이 PI4K가 다양한 암에서 과발현 되어 있음이 보고되었고 이에 따라 PI4K가 항암치료의 잠재적인 표적이 될 수 있음이 제시되고 있다. C형 간염 바이러스 (HCV)가 복제를 위하여 숙주세포의 PI4K의 동종효소 중 하나인 PI4K IIIα를 이용하며, 또한 HCV의 항바이러스제 중 하나인, 4-Anilino Quinazoline이 PI4K IIIα를 억제 함으로서 항바이러스 효과를 나타낸다는 사실이 알려졌다.

이에 최근의 연구에서 본 연구진은 다양한 HCV 항바이러스제를 스크리닝하여 시메프레비르(simeprevir)가 PI4K IIIα의 활성을 억제하는 기능이 있음을 밝혔다. 시메프레비르는 PLC/PKC 및 PI3K/Akt 경로를 모두 억제하여 다양한 인간 암세포에서 방사선민감도를 증가시켰다. 한편 최근 programmed death-1 (PD- 1)/programmed death-ligand 1 (PD-L1)이 면역치료의 중요한 표적임이 밝혀졌고, PD-L1 경로의 조절에 PI3K가 관련이 있음이 제시된 바 있다. PI3K의 다양한 동종효소 중 PI3Kδ가 면역 조절 기능이 있음이 알려졌고 이에 혈액암에서 암치료의 타겟으로 연구되어 왔다. 또한 최근의 연구들에서는 PI3Kδ가 고형암에서 과발현되는 현상 및 PI3Kδ의 억제가 고형암세포에서 항암효과를 나타내는 현상이 보고되었다. 이러한 이전의 연구 결과들에 기반하여, 본 연구에서는 종양 이종이식 모델(tumor xenograft model)에서 방사선 감작 효과를

(39)

확인하고, 그 작용기전을 연구하고자 하였다. 또한 면역능 동계 종양 모델(immune-competent syngeneic murine tumor model)에서 PI4K IIIα의 면역 조절 효과를 평가하였다.

방법: 생체 내 방사선 감작 효과를 확인하기 위하여, 유방암 및 뇌종양 이종이식 모델을 형성하였다. BALB/c 무흉선 누드마우스의 뒷다리에 BT474세포를 이식하였고, 뇌의 꼬리엽 및 조가비핵에 U251세포를 이식하였다. 마우스는 대조군, 시메프레비르, 방사선치료, 시메프레비르+방사선치료의 4개 군으로 랜덤배정 하였다.

시메프레비르는 10 mg/kg의 용량으로 복강 내에 주입하였으며, 일주일에 3번씩 2주 동안 처리하였고, 방사선치료는 9 Gy를 3회에 나누어 1주일간 처리하였다. 모든 처치가 끝난 후 일정한 간격으로 종양의 성장을 측정하였다. 유방암 모델에서 종양의 크기는 (길이×너비×높이)/2의 공식을 이용하여 계산하였으며, 뇌종양 모델의 종양의 크기는 생체발광 이미지를 이용하여 측정하였다. PI4K IIIα의 억제와 방사선치료가 상피간엽이행에 미치는 영향을 확인하기 위하여, wound healing assay 및 modified Boyden chamber assay를 시행하여 BT474 및 U251 세포의 이동과 침습을 평가하였다. 유방암 이종이식 모델에서 얻은 종양 조직에서 PI3Kδ의 발현의 변화를 확인하기 위하여 면역조직화학염색을 시행하였다. RNA간섭을 통하여 PI3Kδ의 억제하였을 때 방사선민감도의 변화를 확인하기 위하여 BT474 및 MDA-MB-468 세포에서 집락형성분석을 시행하였다. RNA간섭과 시메프리비르를 통한 PI3Kδ의 억제가 PD-L1의 발현에 미치는 영향은 웨스턴블롯분석에서 평가하였다. 시메프레비르와 PD-L1 억제제의 병용효과를 확인하기 위하여, 인간 T 림프구와 함께 배양된 BT474 및 MDA-MB-468 세포에서 집락형성분석을 시행하였다. 시메프레비르의 생체 내 면역조절 효과를 확인하기 위하여 면역능 BALB/c 마우스에

(40)

4T1 세포를 이식하여 동계 종양 모델을 제작하였다. 마우스는 이종이식 모델에서와 같은 4개의 그룹으로 랜덤배정하였다. 방사선치료는 8Gy씩 3회를 1주일 동안 처리하였고, 시메프레비르는 20mg/kg 용량으로 일주일에 3번, 2주 동안 처리하였다. 종양의 성장은 유방암 이종이식 모델에서 이용된 공식을 이용하여 계산하였다. 동계 종양 모델에서 얻은 조직을 이용하여 유세포분석 및 웨스턴블롯분석을 시행하였다.

결과: 유방암 이종이식 모델에서, 시메프레비르를 통한 PI4K IIIα 억제와 방사선치료가 병합되었을 때 종양의 성장이 각각의 치료를 단독으로 처리한 군들에 비하여 유의하게 억제되는 것을 확인하였다.

뇌종양 이종이식 모델에서 종양의 크기는 시메프레미르와 방사선치료를 함께 처리한 군에서 가장 작았으나, 이 차이는 통계적 유의성은 없었다.

또한 RNA간섭이나 시메프레비르를 이용하여 PI4K IIIα를 억제하였을 때, wound healing assay 및 modified Boyden chamber assay 에서 암세포의 침습과 이주가 억제되었고, 또한 방사선치료와 병합하였을 때 그 억제효과가 더 강화되었다. 면역조직화학염색 결과, 시메프레비르와 방사선치료를 병합한 군에서 PI3Kδ의 발현이 억제되었다. RNA간섭을 이용한 PI3Kδ의 억제 시, BT474 및 MDA-MB-468 유방암세포에서 방사선 민감도가 증가되었으며, PD-L1 과 p-Akt의 발현이 억제되었다.

다양한 뇌종양 및 유방암세포를 스크리닝 하였을 때, U251 세포와 MDA-MB-468 세포에서 시메프레비르가 PD-L1의 발현을 억제함을 확인하였다. BT474 및 MDA-MB-468 유방암세포에서 시메프레비르와 PD-L1 억제제의 병합하였을 때, PD-L1 억제제의 방사선 감작 효과가 증가되었고, p-AKT의 발현이 억제되었다. 면역능 동계 종양 모델에서, 종양의 성장이 시메프레비르 및 방사선치료를 병합한 군에서 유의하게 억제되었다. 동계 종양 모델에서 얻은 조직을

(41)

T 세포의 비율이 현저히 증가하였으며, PI3Kδ의 발현이 억제되었다 결론: 시메프레비르를 이용하여 PI4K IIIα를 억제하였을 때, 이종이식 모델과 면역능 동계 종양 모델 모두에서 유의한 방사선 감작 효과를 확인할 수 있었다. 시메프레비르는 PI3Kδ의 발현을 억제하고 상피간엽이행을 억제하였다. PI4K IIIα의 억제는 PI3Kδ의 발현을 조절하여 PD-L1을 조절하였고, T 세포의 CD4(+)/CD8(+) 비율을 조절 함으로서 면역 조절 효과를 나타내었다. 결론적으로 시메프레비르를 통하여 PI4K IIIα를 억제하는 것은 방사선치료의 효율을 높이고 면역 체크포인트 차단제의 효과를 증가시키는 유용한 전략이 될 수 있을 것으로 생각된다.

--- 주요어: 포스파티딜이노시톨 4-키나아제 IIIα, 방사선민감도, 면역 체크포인트 차단제, 포스파티딜이노시톨 3-키나아제 δ

학 번: 2015-30597

Gambar

Figure  1.  Radiosensitizing  effect  of  simeprevir  in  an  in  vivo  human  breast  tumor xenograft model
Figure  2.  Radiosensitizing  effect  of  simeprevir  in  an  in  vivo  human  brain  tumor  xenograft  model
Figure 3. Effect of PI4K IIIα inhibition on migration and invasion.  Migration  and  invasion  potential  of  U251  cells  and  BT474  cells  were  assessed  with  wound  healing assays (A) and modified Boyden chamber assay (B), respectively
Figure  5.  Radiosensitizing  effect  of  PI3Kδ  inhibition  using  RNAi.  Specific  inhibition  of  PI3Kδ  using  RNA  interference  (RNAi) increased  radiosensitivity  of  BT474 and MDA-MB-468 cells
+5

Referensi

Dokumen terkait

Wiegand 등의 연구에서는, bovine dentin을 이용한 실험에서 다양한 전동칫솔을 이용한 마모도 실험결과, 부식된 상아질과 건전한 상아질 모두 수동칫솔보다 전동칫솔에서 더 높 은 마모도를 보였다.12 하지만, 아직까지 연마제의 함량이 다양한 시 판된 세치제를 이용하여 이에 따른 전동칫솔과 수동칫솔의 상아질 마모 도

EBSD orientation maps ND and inverse pole figure for recrystallized grains obtained from electropulsing treated specimens for target temperature of 760℃ with current density of a

GNSS 위치 정보 기반의 방위각 데이터를 이용하여 노선별 상·하행 데이터를 분리하 고, 시간평균속도와 공간평균속도를 이용한 공간의 점진적 분리를 통한 속도 동질공간을 결정하였다.. 경위도 각각 2개로 분할하는 4진법 기준의 분할 방법을 적용한 지오해시를 이용하여 대용량 데이터의 처리속도를

인출 전략 의 경우 기존 선행연구들에서 인출 전략을 활용하는 것이 학습에 효과적 일 수 있다는 것이 보고되었으며 초등학생들에게서도 인출 전략의 효과 를 보고한 선행연구들이 있었지만Karpicke & Blunt, 2011; Karpicke et al., 2016 본 연구의 결과에 따르면 실제로 초등학생의 학습전략 활용에 있어서

그리고 예술에 있어서 이상향에 대한 노스탤지어의 대 표적 유형과 역사적 전개를 연구자의 작품 속 이상향의 노스탤지어가 담 긴 지평 표현 및 변천 과정과 비교함으로써 작품에 내재한 이상향의 본 질과 인식의 문제를 규명하는 데 연구의 목적을 두었다.. 결론’의 여섯 부분으로 구성되어

선별적 패킷 전송 지연 기법은 지연 전송이 허용되는 패킷의 전송 요청이 발생했을 때, 해당 패킷의 전송 지연이 모뎀의 전력 소모량을 줄일 것으로 예상될 때에만 실제로 전송을 지연시키는 기법이다... 구체적으로, 이 모델은 패킷 전송 요청이 발생하는 시점의 모뎀의 정확한 상태와, 패킷 전송을 지연했을 때 예상되는 모뎀의 향후 상태

TABLES [Table 1] Demographic characteristics of pilot survey subjects ··· 23 [Table 2] Descriptive statistics about collective efficacy questions ··· 25 [Table 3] Exploratory

이 연구에서는 제2자 처벌 게임과 제3자 처벌 게임에서 자기애 성향이 높은 피험자가 분배자의 돈을 감소시키기 위해 얼마나 많은 돈을 쓰는지를 알아보았고 두 실험의 처벌 정도를 평균 내어 자기애가 낮은 집단보다 자기애 집단이 처벌에 더 많은 비용을 지불하는 것을 확인했다.. 제2자 처벌 게임과 제3자 처벌 게임에서 사회적 규범을