Categories
Articles

Knockdown of insulin-like growth factor 1 receptor enhances chemosensitivity to cisplatin in human lung adenocarcinoma A549 cells

Original Paper

Pdf

file on Synergy OPEN

omments

Acta Biochim Biophys

Sin 2008, 40: 497-504

doi:10.1111/j.1745-7270.2008.00429.x

Knockdown of insulin-like

growth factor 1 receptor enhances chemosensitivity to cisplatin in human lung

adenocarcinoma A549 cells

Aiqiang Dong1,

Minjian Kong1, Zhiyuan Ma2,

Jianfang Qian1, Haifeng Cheng1,

and Xiaohong Xu3*

1 Department of Cardiothoracic Surgery, Second

Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310009,

China

2 Department of Thoracic and Cardiovascular

Surgery, Shanghai Jiao Tong University Affiliated First People’s Hospital,

Shanghai 200080, China

3 Department of Endocrinology, Second Affiliated

Hospital, School of Medicine, Zhejiang University, Hangzhou 310009, China

Received: March 24,

2008       

Accepted: May 6,

2008

This work was

supported by a grant from the natural

Science Foundation of Zhejiang Province (Y-204317)

*Corresponding

author: Tel, 86-571-87783516; fax,

86-571-87022660; E-mail, [email protected]

The effects

of RNA interference-mediated insulin-like growth factor 1 receptor (IGF1R) gene

silencing in response to cisplatin (DDP) in the lung cancer cell line A549 in

vivo and in vitro were investigated using two plasmids expressing

short hairpin RNA (shRNA) to IGF1R. A549 cells were transfected with plasmids

expressing each shRNA and then treated with DDP. Semi-quantitative reverse

transcription-PCR and Western blot analysis were used to detect the expression

of IGF1R. MTT assay, flow cytometry and tumor growth assay in athymic nude mice

were used to assess the chemosensitivity to DDP following IGF1R knockdown. Our

data showed that the transfection of A549 cells with shRNA resulted in specific

silencing of IGF1R by 78.9% at the mRNA level and by 89.8% at the protein

level. Down-regulation of IGF1R significantly enhanced cell sensitivity to DDP,

decreased the IC50 of DDP in

A549 cells at 24 h, 48 h and 72 h, and retained 77.5% of A549 cells in the G0/G1 phase.

Furthermore, shRNA-mediated silencing of IGF1R in combination with DDP

treatment enhanced the suppression of tumor growth in both size and weight

by more than 60% and increased apoptosis by more than 75% when compared with

the controls in vivo. Suppression of IGF1R gene expression by shRNA

enhances the chemosensitivity of A549 cells to DDP both in vitro and in

vivo, indicating the therapeutic potential of RNA interference as a method

for gene therapy in treating lung cancer.

Keywords    lung cancer; insulin-like growth factor 1 receptor; RNA

interference; cisplatin; chemosensitivity

Lung cancer is the leading cause of cancer death worldwide,

accounting for 32% of cancer deaths in men and 24% in women. Non-small cell

lung cancer (NSCLC) comprises approximately 75%80% of lung cancers, and

the overall 5-year survival rate for NSCLC is only 8%14% when diagnosed and 40%

after complete surgical resection [1,2]. Meta-analysis reveals that combined

chemotherapy based on cisplastin (DDP) is the standard therapy for NSCLC; it is

recommended by the American Society of Clinical Oncology (ASCO) for patients

with advanced stage NSCLC. However, the efficacy of this treatment is limited,

and the 5-year survival rate is only 20%40% [3], which may be due

to the multiple drug resistance of NSCLC cells to chemical agents. The

development of novel strategies for enhancing chemosensitivity is the focus of

much medical research.Insulin-like growth factor 1 receptor (IGF1R; GenBank accession No.

NM_000875) is a receptor protein tyrosine kinase, which is a transmembrane

heterotetramer consisting of two a-subunits and two b-subunits linked by

disulfide bonds. Binding of ligands to the receptor leads to receptor

oligomerization, activation of protein tyrosine kinase, intermolecular receptor

autophosphorylation and phosphorylation of cellular substrates that consequently

cause gene activation, DNA synthesis and cell proliferation [48]. Recent

studies have shown that IGF1R is overexpressed in various human cancers,

including lung cancer [9], and that overexpression or constitutive activation

of IGF1R results in ligand-dependent transformation of fibroblasts [10]. IGF1R

also plays a key role in the survival of transformed colonocytes and leads to

the development of tumors in nude mice [11,12]. Besides its role in

tumorigenesis, IGF1R protects tumor cells from apoptosis induced by

chemotherapy, radiotherapy or cytokine [1315]. In our previous studies [1618], we constructed IGF1R-specific short

hairpin RNA (shRNA)-expressing plasmids and transfected A549 cells. It was found

that the proliferation, adhesion and invasion of A549 cells were inhibited. In

this study, we further detect the combined effect of IGF1R-specific shRNA with

DDP in A549 cells.

Material and Methods

Materials

Anti-IGF1R monoclonal antibody (MS-645-P0) was purchased from Lab

Vision (New York, USA). Anti-human actin polyclonal antibody (sc-1616),

horseradish peroxidase (HRP)-conjugated goat anti-mouse (sc-2060) and goat

anti-rabbit IgG-HRP IgG (sc-2004) antibodies were purchased from Santa Cruz

Biotechnology (Santa Cruz, USA). BLOCK-iTTM U6 RNA interference (RNAi) entry

vector kit was purchased from Invitrogen (Carlsbad, USA). AxyPrep Multisource

Genomic DNA Miniprep kit was purchased from Axygen Biosciences (Union City,

USA). Specific pathogen-free male BALB/cAnNCrj-nu mice (six weeks old) were

purchased from the Shanghai Cancer Institute (Shanghai, China). The mice were

cared for and used according to Zhejiang University’s guidelines.

Construction of plasmids

expressing shRNA targeting IGF1R

Plasmids expressing two shRNAs targeting IGF1R were constructed and

designated IGF1R-shRNA1 and IGF1R-shRNA2, respectively [16]. A plasmid

expressing shRNA targeting the Photinus pyralis luciferase gene (X65324)

was used as a control (control shRNA). The shRNA sequences contained in the

IGF1R-shRNA1, IGF1R-shRNA2 and control shRNA were as follows: 5-CAC­C­G­CACA­ATTAC­TGCTCCAAAGACG­AATCTTTG­GAGCAGTAA­TTG­TGC-3,

5-CACCGCCGA­TGTGTGAGAAG­ACCT­T­C­­­­A­­A­GAGAGGTCTTCTCACACATCGGC-3 and 5-CA­C­CGCTCACCGGCTCCAGATTTATCGAAA­TAAAT­CT­G­G­­AGCCGGTGAGC-3.

Cell culture and transfection

Human lung adenocarcinoma A549 cells were cultured in RPMI 1640

medium supplemented with 10% fetal bovine serum (FBS) at 37 ?C with 5% CO2. For transfection, 2 ml of A549 cells was seeded in a 6-well plate

at the concentration of 3?105 cells/ml for 16 h. Cells were then transfected with a mixture of 4 mg of each

plasmids and 10 ml of Lipofectamine 2000 (Invitrogen) in 2 ml serum-free medium. Six

hours after transfection, the medium was replaced with fresh RPMI 1640 medium

supplemented with 10% FBS and cultured for 48 h.

Reverse

transcription-polymerase chain reaction (RT-PCR) and Western blot analysis

Total cellular RNA and proteins were prepared as described

previously [16]. PCR primer sequences for IGF1R were forward

5-AAATGTGCCCGAGCGTGTG-3 and reverse 5-TGCCCTTGAAGATGGTGCATC-3, and for human b-actin, the

internal control, they were forward 5-TTCCA­GCCTTCCTTCCTGGG-3 and reverse

5-TTGCGC­TC­A­G­GAGGAGCATT-3. The thermal cycle conditions were: 94 ?C for 3

min followed by 35 cycles for IGF1R (25 cycles for b-actin) at 94 ?C for 30 s,

53.5 ?C for 30 s, 72 ?C for 60 s, and a final extension at 72 ?C for 10 min.

The PCR product (10 ml) was electrophoresed on 2% agarose, stained with ethidium bromide

and visualized by UV absorption. Total cellular RNA and proteins were prepared as described

previously [16]. PCR primer sequences for IGF1R were forward

5-AAATGTGCCCGAGCGTGTG-3 and reverse 5-TGCCCTTGAAGATGGTGCATC-3, and for human b-actin, the

internal control, they were forward 5-TTCCA­GCCTTCCTTCCTGGG-3 and reverse

5-TTGCGC­TC­A­G­GAGGAGCATT-3. The thermal cycle conditions were: 94 ?C for 3

min followed by 35 cycles for IGF1R (25 cycles for b-actin) at 94 ?C for 30 s,

53.5 ?C for 30 s, 72 ?C for 60 s, and a final extension at 72 ?C for 10 min.

The PCR product (10 ml) was electrophoresed on 2% agarose, stained with ethidium bromide

and visualized by UV absorption. For Western

blot analysis, 100 mg of total cellular proteins (30 mg for b-actin)

were separated on 10% SDS-polyacrylamide gels, and then transferred to Hybond-P

polyvinylidene difluoride (PVDF) membranes (Amersham, Piscataway, USA). After

the non-specific binding sites were blocked by incubating the membranes in

phosphate-buffered saline-0.1% Tween-20 (PBS-T) without skimmed milk

(Tris-buffered saline-0.1% Tween-20 (TBS-T) with 5% skimmed milk for b-actin) for

0.5 h at room temperature, the membranes were probed with primary antibodies

(1:200 dilution for IGF1R for 2 h; 1:1000 dilution for b-actin for 3 h) at

room temperature, and then washed three times with PBS-T (TBS-T for b-actin).

The membranes were then incubated with HRP-conjugated goat anti-mouse (or goat

anti-rabbit) IgG (1:2500 dilution) for 1 h at room temperature, and washed

three times with PBS-T (TBS-T for b-actin). The blots were developed by chemiluminescence kit

(Roche, Indianapolis, USA), and analyzed using Scion Image software (Scion

Corporation, Frederick, USA).

Assessment of the effect on

the chemosensitivity to DDP by MTT assay

A549 cells were seeded in a 96-well plate at a concentration of 4?103 cells/well for 16 h and then transfected with

IGF1R-shRNA1, control shRNA or PBS as the negative control. The medium was

changed with RPMI 1640 medium 6 h later. Then, DDP was added to every 4 wells

at the concentration of 0.1, 0.5, 1, 2, 4 and 8 mg/L. Cell viability was

measured at 24 h, 48 h and 72 h by MTT assay at 570 nm (OD readings) after DDP

or PBS treatment. The suppression rate was calculated using the formula:

Eq.

The suppression rate curve at different DDP concentrations was made

to calculate the IC50 using curve regression. All the experiments

were performed in triplicate.

Evaluation of the effect of

DDP treatment on cell cycle by flow cytometry

A549 cells were seeded into 6-well plates for 16 h and then

transfected with IGF1R-shRNA1 or control shRNA. The medium was replaced with

RPMI 1640 medium 6 h later. Then, 0.5 mg/L DDP was added into each group. PBS

was used as the negative control. Cells were removed by trypsinization 48 h after

the addition of DDP, washed in PBS and fixed in 70% ice-cold ethanol for 2 h.

The fixed samples were centrifuged, treated with 1 mg/ml RNase solution (Sigma,

St. Louis, USA) for 30 min at 37 ?C, and resuspended in 0.1 mg/ml propidium

iodide (PI) solution (Sigma) at 4 ?C for 1 h. PI-stained cells were analyzed

with a flow cytometer (BD Biosciences, San Jose, USA). Cell cycle and apoptotic

rate were analyzed by CellQuest 3.1f and ModFit 3.0 DNA software (BD

Biosciences).

Measurement of the effect on tumorigenicity

in vivo

To assess the effect of IGF1R-shRNA1 on chemotherapy in

tumorigenicity, 12 six-week-old, male nude mice were randomly divided into

three groups. A549 cells transfected with IGF1R-shRNA1 or control shRNA or

treated with PBS were subcutaneously inoculated into murine back region at a

concentration of 5?107

cells/mouse. On day 2, mice in the IGF1R-shRNA1 and control shRNA groups were

administrated i.p. with 100 ml of 1 mg/L DDP twice a day for 10 d while the mice in the

PBS-treated group were administrated i.p. with 100 ml PBS twice a day. Tumor

size was measured every 5 d and calculated by the formula:

Eq.

After 30 d, the mice were euthanized. The tumors were removed, fixed

by 4% polyformaldehyde, paraffin embedded and sectioned. Formalin-fixed

paraffin sections were used for terminal deoxynucleotidyl transferase-mediated

digoxigenin-dUTP nick-end labeling (TUNEL) assay. The numbers of apoptotic

cells in tumor tissue on each section were counted in 10 different microscopic

fields. Proteins were extracted from 50 mg tumor tissue, and Western blotting

was used to analyze the expression of IGF1R.

Statistical analysis

All the quantitative data were

presented as mean±SD. The statistical significance of the differences was

determined using Student’s two-tailed t-test for two groups and one-way

ANOVA for multiple groups. A P-value less than 0.05 was considered

statistically significant. All the data were analyzed with the SPSS 13.0

software (SPSS, Chicago, USA).

Results

Transfection of shRNA

suppresses IGF1R expression in A549 cells

To inhibit IGF1R gene expression with shRNA, we constructed two

plasmids expressing shRNA to IGF1R. RT-PCR and Western blot analysis were

performed. The results showed that IGF1R mRNA expression in cells

transfected with IGF1R-shRNA1 was 21.1%3.5% of that for those transfected with

control shRNA [P<0.05; Fig. 1(A)]. Similarly, immunoblot

analysis revealed that the expression of IGF1R protein in A549 cells

transfected with IGF1R-shRNA1 was strongly inhibited. Densitometric analysis

showed that the amounts of IGF1R protein remaining in cells transfected with

IGF1R-shRNA1 and IGF1R-shRNA2 was 10.2%2.8% and 47.9%15.8%, respectively, of

that found in cells transfected with the plasmid control shRNA [P<0.05; Fig. 1(B)]. These results indicated that the transfection of shRNA can

effectively suppress IGF1R expression. IGF1R-shRNA1 proved to be more potent

than IGF1R-shRNA2, so we used IGF1R-shRNA1 in the subsequent experiments.

IGF1R gene silencing sensitizes

A549 cells to chemotherapy

To investigate whether IGF1R down-regulation alters chemosensitivity

in A549 cells, we tested DDP in cells transfected with shRNA against IGF1R. The

cell growth suppression was measured at 24 h, 48 h, and 72 h by MTT assay, as

shown in Fig. 2. The suppression rate in cells transfected with

IGF1R-shRNA1, particularly in those treated with 0.5 to 4 mg/L DDP, was

significantly higher than those transfected with control shRNA. IC50 at different time points was also markedly decreased (Table 1).

Suppression of IGF1R

expression blocks A549 cells at G0/G1 and increases apoptosis when

combined with DDP

To determine the apoptosis-inducing potential of IGF1R-shRNA1 in A549

cells, flow cytometric analysis of PI-stained cells was performed. As indicated

in the previous study [16], the percentage of cells at the G0/G1 population after transfection with IGF1R-shRNA1

(77.5%) was much higher than that observed after transfection with control

shRNA (47.2%), and those at S phase and G2/M phase

were 15.7% and 7.3% in IGF1R-shRNA1 group, lower than the 23.0% and 29.9% in

the control, respectively. The apoptotic rate in cells transfected with

IGF1R-shRNA1 combined with DDP (0.5 mg/L) was 44.2%, much higher than that

(27.8%) in the control (Fig. 3).

Combination of IGF1R

gene silencing and chemotherapy inhibits in vivo tumorigenicity

We evaluated the effects of receptor blockade on chemotherapy in

vivo. The incidence of subcutaneous tumors derived from A549 cells were

100%. The time for tumorigenicity was 20 d for the group treated with

IGF1R-shRNA1 and DDP, 20 d for the group treated with control shRNA and DDP, and

15 d for the PBS group. All mice survived for 30 d after inoculation, and the

tumor growth rate in the IGF1R-shRNA1 group was much lower than that in the

other two groups (P<0.05). The tumor size and weight in the group treated with IGF1R-shRNA1 and DDP were 20.74.2 mm3 and

70.012.0 mg, which was markedly lower than the tumor size (50.315.2 mm3) and weight (180.020.0 mg) in the group treated with control shRNA

and DDP (P<0.05; Table 2). The tumors in the group treated

with IGF1R-shRNA1 and DDP were oval and had a smooth surface while the tumors

in the other two groups were irregular, nodular and rich in vessels (Fig. 4).

There was no invasion in any of the groups. These results suggested that the

combination of IGF1R gene silencing and chemotherapy could significantly

inhibit the tumor growth of A549 cells in nude mice.

Suppression of IGF1R combined

with DDP treatment increases cell apoptosis and decreases IGF1R expression in

established tumors

TUNEL assay was further performed to evaluate apoptosis of tumor

tissues in vivo. The number of apoptotic cells was significantly

increased in the group treated with IGF1R-shRNA1 and DDP (141.39.1) when

compared with the group treated with control shRNA and DDP (34.57.6) (P<0.05; Fig. 5). Western blot analysis showed significantly decreased amounts of

IGF1R protein in the group treated with IGF1R-shRNA1 and DDP when compared with

the group treated control shRNA and DDP and the PBS group (Fig. 6).

These data indicated that shRNA-mediated IGF1R gene silence combined with

chemotherapy may also induce apoptosis of A549 cells in vivo.

Discussion

It has been reported that IGF1R mediates tumor growth and protects

cancer cells from apoptosis [1921]. In order to blunt IGF1R function or expression, a number of experimental

strategies have been employed. For instance, a-IR3, a monoclonal antibody

to IGF1R which blocks IGF1R signaling, significantly inhibits Ewing’s sarcoma

cells in vitro and induces the regression of established tumors [22].

Genetic blockage can be accomplished using an antisense oligonucleotide [23],

and vectors [24] expressing antisense IGF1R mRNA have been shown to inhibit

cell growth, suppress tumorigenesis, alter the metastatic potential and prolong

survival in vivo. Another approach is to utilize dominant-negative

mutants to inhibit the function of the naturally expressed receptor rather than

its expression. Using mutant receptors for IGF1R that contain a portion of the

molecule including only the extracellular domain or the extracellular domain

with a mutant or deleted intracellular tyrosine kinase domain induces

differentiation and inhibits adhesion, invasion and metastasis [2526]. In this

study, we constructed two plasmids expressing shRNA to IGF1R under the control

of the human U6 promoter, IGF1R-shRNA1 and 2, and then evaluated the effects of

shRNA on IGF1R expression in A549 cells. Our results show that IGF1R expression

significantly inhibited in A549 cells at both mRNA and protein levels by shRNA

to IGF1R [27], suggesting that vector-based RNAi may be a potential approach to

cancer gene therapy.It has also been reported that IGF1R signaling results in

chemotherapy resistance in a wide variety of tumors. For example, blockage of

IGF1R signaling using a monoclonal antibody or RNAi enhances sensitivity to

chemotherapy in breast and liver cancer cell lines [28,29]. An anti-IGF1R

antibody enhances the chemosensitivity to gemcitabine in the pancreatic cancer

xenografts [30], and an antisense inhibition of IGF1R increases sensitivity of

prostate cancer cells to cisplatin, mitoxantrone and paclitaxel [31].

In agreement with these results, our study shows that knockdown of

IGF1R by shRNA alters chemosensitivity and results in a significant increase in

sensitivity to DDP. Specifically, IGF1R-shRNA1 combined with DDP treatment in

A549 cells significantly decreases IC50, arrests A549 cells at

G0/G1 and increases apoptosis in vitro.

Similarly, suppression of IGF1R combined with DDP treatment inhibits in vivo

tumorigenicity, increases cell apoptosis and decreases IGF1R expression in

established tumors.Taken together, our findings indicate that IGF1R expression can be

inhibited by shRNA to IGF1R, and shRNA-mediated silencing of IGF1R results in

sensitization to DDP in lung cancer A549 cells both in vitro and in

vivo. Our results provide further evidence that IGF1R targeting is a

potential therapy for human lung cancer.

References

 1   Mezzetti M, Panigalli T, Giuliani L, Raveglia

F, Giudice FL, Meda S. Personal experience in lung cancer sleeve lobectomy and

sleeve pneumonectomy. Ann Thorac Surg 2002, 73: 17361739

 2   Greenlee RT, Murray T, Bolden S, Wingo PA.

Cancer statistics. CA Cancer J Clin 2000, 50: 733

 3   Bunn PA Jr, Kelly K. New combinations in the

treatment of lung cancer: a time for optimism. Chest 2000, 117: 138143

 4   Yarden Y, Ullrich A. Growth factor receptor

tyrosine kinases. Annu Rev Biochem 1988, 57: 443478

 5   Williams LT. Signal transduction by the

platelet-derived growth factor receptor. Science 1989, 243: 15641570

 6   Cantley LC, Auger KR, Carpenter C, Duckworth

B, Graziani A, Kapeller R. Oncogenes and signal transduction. Cell 1991, 64:

281302

 7   Schlessinger J. Signal transduction by

allosteric receptor oligomerization. Trends Biochem Sci 1988, 13: 443447

 8   Yarden Y, Schlessinger J. Epidermal growth

factor induces rapid, reversible aggregation of the purified epidermal growth

factor receptor. Biochemistry 1987, 26: 14431451

 9   LeRoith D, Werner H, Beitner-Johnson D,

Roberts CT Jr. Molecular and cellular aspects of the insulin-like growth factor

I receptor. Endocr Rev 1995, 16: 143163

10  Kaleko M, Rutter WJ, Miller AD. Overexpression

of the human insulinlike growth factor I receptor promotes ligand-dependent

neoplastic transformation. Mol Cell Biol 1990, 10: 464473

11  Remacle-Bonnet MM, Garrouste FL, Heller S,

Andre F, Marvaldi JL, Pommier GJ. Insulin-like growth factor-I protects colon

cancer cells from death factor-induced apoptosis by potentiating tumor necrosis

factor alpha-induced mitogen-activated protein kinase and nuclear factor kappaB

signaling pathways. Cancer Res 2000, 60: 20072017

12  Singh P. Insulin-like growth factor system in

growth, development and carcinogenesis. J Clin Lig Assay 2000, 23: 214232

13  Scotlandi K, Maini C, Manara MC, Benini S,

Serra M, Cerisano V, Strammiello R et al. Effectiveness of insulin-like

growth factor I receptor antisense strategy against Ewing’s sarcoma cells.

Cancer Gene Ther 2002, 9: 296307

14  Turner BC, Haffty BG, Narayanan L, Yuan J,

Havre PA, Gumbs AA, Kaplan L et al. Insulin-like growth factor-I

receptor overexpression mediates cellular radioresistance and local breast

cancer recurrence after lumpectomy and radiation. Cancer Res 1997, 57: 30793083

15  Wu Y, Tewari M, Cui S, Rubin R. Activation of

the insulin-like growth factor-I receptor inhibits tumor necrosis

factor-induced cell death. J Cell Physiol 1996, 168: 499509

16  Dong AQ, Kong MJ, Ma ZY, Qian JF, Xu XH.

Down-regulation of IGF-IR using small, interfering, hairpin RNA (siRNA)

inhibits growth of human lung cancer cell line A549 in vitro and in nude

mice. Cell Biol Int 2007, 31: 500507

17  Qian J, Dong A, Kong M, Ma Z, Fan J, Jiang G.

Suppression of type 1 Insulin-like growth factor receptor expression by small

interfering RNA inhibits A549 human lung cancer cell invasion in vitro

and metastasis in xenograft nude mice. Acta Biochim Biophys Sin 2007, 39: 137147

18  Dong AQ, Kong MJ, Ma ZY, Qian JF, Fan JQ, Xu

XH. shRNA-mediated insulin-like growth factor I receptor gene silencing

inhibits cell proliferation, induces cell apoptosis, and suppresses tumor

growth in non-small cell lung cancer: in vitro and in vivo experiments.

Zhonghua Yi Xue Za Zhi 2007, 87: 15061509

19  Kulik G, Klippel A, Weber MJ. Antiapoptotic

signaling by the insulin-like growth factor I receptor, phosphatidylinositol

3-kinase, and Akt. Mol Cell Biol 1997, 17: 15951606

20  Rochester MA, Riedemann J, Hellawell GO, Brewster

SF, Macaulay VM. Silencing of the IGF1R gene enhances sensitivity to

DNA-damaging agents in both PTEN wild-type and mutant human prostate cancer.

Cancer Gene Ther 2005, 12: 90100

21  Ma J, Pollak MN, Giovannucci E, Chan JM, Tao

Y, Hennekens CH. Prospective study of colorectal cancer risk in men and plasma

levels of insulin-like growth factor (IGF)-I and IGF-binding protein-3. J Natl

Cancer Inst 1999, 91: 620625

22  Scotlandi K, Benini S, Nanni P, Lollini PL,

Nicoletti G, Landuzzi L, Serra M et al. Blockage of insulin-like growth

factor-I receptor inhibits the growth of Ewing’s sarcoma in athymic mice.

Cancer Res 1998, 58: 41274131

23  Chernicky CL, Yi L, Tan H, Gan SU, Ilan J.

Treatment of human breast cancer cells with antisense RNA to the type I insulin-like

growth factor receptor inhibits cell growth, suppresses tumorigenesis, alters

the metastatic potential, and prolongs survival in vivo. Cancer Gene

Ther 2000, 7: 384395

24  Lee CT, Wu S, Gabrilovich D, Chen H,

Nadaf-Rahrov S, Ciernik IF, Carbone DP. Antitumor effects of an adenovirus

expressing antisense insulin-like growth factor I receptor on human lung cancer

cell lines. Cancer Res 1996, 56: 30383041

25  Yuen JS, Macaulay VM. Targeting the type 1

insulin-like growth factor receptor as a treatment for cancer. Expert Opin Ther

Targets 2008, 12: 589603

26  Lee CT, Park KH, Adachi Y, Seol JY, Yoo CG,

Kim YW, Han SK et al. Recombinant adenoviruses expressing dominant

negative insulin-like growth factor-I receptor demonstrate anti-tumor effects

on lung cancer. Cancer Gene Ther 2003, 10: 5763

27  Zhang H, Pelzer AM, Kiang DT, Yee D.

Down-regulation of type I insulin-like growth factor receptor increases

sensitivity of breast cancer cells to insulin. Cancer Res 2007, 67: 391397

28  Beech DJ, Parekh N, Pang Y. Insulin-like

growth factor-I receptor antagonism results in increased cytotoxicity of breast

cancer cells to doxorubicin and taxol. Oncol Rep 2001, 8: 325329

29  Niu J, Xu Z, Li XN, Han Z. siRNA-mediated type

1 insulin-like growth factor receptor silencing induces chemosensitization of a

human liver cancer cell line with mutant P53. Cell Biol Int 2007, 31: 156164

30  Maloney EK, McLaughlin JL, Dagdigian NE

Garrett LM, Connors KM, Zhou XM, Blattler WA et al. An anti-insulin-like

growth factor I receptor antibody that is a potent inhibitor of cancer cell

proliferation. Cancer Res 2003, 63: 50735083

31     Hellawell GO, Ferguson DJ, Brewster SF,

Macaulay VM. Chemosensitization of human prostate cancer using antisense agents

targeting the type 1 insulin-like growth factor receptor. Br J Urol 2003, 91:

271277