Korean J Intern Med > Volume 17(3); 2002 > Article
Joo, Sohn, Lee, Park, Choi, Rew, Park, and Kim: Expression of Vascular Endothelial Growth Factor and p53 in Pancreatic Carcinomas

Abstract

Background:

Angiogenesis has been shown to be a critical aspect of tumor growth and progression. Vascular endothelial growth factor (VEGF) has potent angiogenic activity and has been identified in a wide variety of malignancies, including pancreatic carcinoma. The tumor-suppressor gene p53 has been thought to regulate VEGF in angiogenesis. The aim of the current study was conducted to investigate the association between p53 mutation and VEGF expression and the prognostic value of these factors in pancreatic carcinoma.

Methods :

Formalin-fixed, paraffin-embedded tissue specimens were obtained from 30 patients who underwent surgery for pancreatic carcinoma. We used an immunohistochemical technique to localize VEGF and p53 in pancreatic carcinoma tissues.

Results :

Positive expression of VEGF was detected in 17 out of 30 (56.7%) tumors. Positive expression of VEGF correlated with the depth of tumor invasion (p=0.002). There was a trend towards an association between positive expression of VEGF and distant metastasis, although these associations were not statistically significant (p=0.070). p53 mutations were identified in 18 out of 30 (60.0%) tumors. However, no significant correlation was found between p53 expression and various clinicopathological parameters. The correlation between p53 mutation and VEGF expression was statistically significant (p=0.004).

Conclusion :

VEGF, a key factor for the induction of tumor-associated angiogenesis, may be involved in tumor characteristics, including tumor invasion and metastasis. And p53 mutation may be implicated in the regulation of angiogenesis through a VEGF up-regulation.

INTRODUCTION

The incidence of pancreatic carcinoma has risen steadily over the recent decades. Since pancreatic carcinoma is diagnosed at an advanced stage and because of the lack of effective therapies, the prognosis of such patients is extremely poor. Surgery being the only potential cure, the prognosis of these patients is critically determined by diagnosing early stage pancreatic carcinoma1,2).
Angiogenesis is an essential process for the primary tumor to grow and invade into the adjacent normal structures. Angiogenesis is likewise considered to permit shedding of cells from a primary tumor to distant body sites, thus facilitating the metastatic process35). The induction of angiogenesis by a tumor is a controlled process, influenced by angiogenic and angiostatic regulators which involve a complex interaction between tumor and endothelial cells6,7). Among the many reported angiogenic regulators, vascular endothelial growth factor (VEGF) is an endothelial cell-specific mitogen and an angiogenic regulator released by a variety of tumor cells810). Expression of VEGF is increased in various human tumors compared with normal tissues, often correlating with tumor angiogenesis and poor prognosis1116). Also, similar studies have shown these findings in pancreatic carcinoma1725).
Recognition of the importance of angiogenesis for the growth and metastasis of cancers has raised fundamental questions regarding the molecular mechanisms of the angiogenic switch during tumor progression. The genetic alterations that are responsible for oncogenesis and tumor progression may underlie the ability of tumors to switch to an angiogenic phenotype6,7). Mutations of the p53 tumor suppressor gene represent one of the most common genetic alterations in human cancers and the acquisition of such defects is strongly associated with tumor progression and metastasis2629). Recently, expression of the p53 gene has been demonstrated to be related to tumor angiogenesis3034). A mutant p53 gene has been suggested to contribute to the angiogenic process through suppression of a strong inhibitor of angiogenesis, namely thrombospondine-1. Evidence also suggests that a mutant p53 gene is a potent stimulant of VEGF.
A few studies have indicated that angiogenesis may be regulated, in part, by function of the p53 tumor suppressor gene in human pancreatic carcinoma18,24). The aim of the current study was conducted to investigate the association between p53 mutation and VEGF expression, and the relationship between these factors and various clinicopathological parameters in pancreatic carcinoma.

MATERIALS and METHODS

Clinical Materials

The specimens of pancreatic carcinomas were obtained from 30 patients who underwent surgery from January 1998 to December 2000 at Chonnam National University Hospital, Gwangju, Korea. None of the patients had undergone chemotherapy or radiotherapy before surgery. Formalin-fixed and paraffin-embedded tissue specimens were taken from representative cancerous lesions over their greatest length and included the adjacent noncancerous area. The clinicopathological findings, including sex, age, tumor size, tumor location, histologic grade and stage, were obtained by medical records and pathologist and physician contact when necessary. The tumors were classified by stages according to the American Joint Committee on Cancer35) and by histological types according to Social et al36). This study group comprised 20 males and 10 females. The mean age was 59.8±12.0 (mean+SD) with a range from 30 to 81 years. The mean size of the tumor was 4.3±1.6 (mean±SD) with a range from 2.0 to 7.5 cm.

Immunohistochemistry

All procedures for immunohistochemical staining were done by the Micro-Probe staining system (Fisher Scientific, Pittsburgh, PA) based on capillary action37). Paraffin sections, of 5 μm in thickness with mounted probe on slides, were immunostained with anti-mouse monoclonal antibodies by the avidin-biotin peroxidase complex method37). Three serial sections of each specimen were stained. The immunostaining was considered to be positive only if the color reaction was equal in intensity and pattern for each section. Sections were deparaffinized and heated in a microwave oven for 7 minutes to retrieve the antigens. They were immersed in 0.6% hydrogen peroxide for 5 minutes to block the endogenous peroxidase activity. A polyclonal antibody against VEGF (A-20; diluted 1:100; Santa Cruz Biotechnology, Santa Cruz, Calf.) and a monoclonal mouse antihuman p53 antibody (DO-7, diluted 1:150; Dakopatts, Glostrup, Denmark) were used as primary antibodies. The primary antibodies, in the aforementioned concentrations were diluted in phosphate buffered saline supplemented with 5% normal horse serum and 1% bovine serum albumin and then incubated with tissues for 120 minutes at room temperature. Anti-mouse immunoglobulin G (Sigma, St. Louis, MO) labeled with biotin was added as a secondary antibody for the detection of primary antibodies and the samples were incubated for 10 minutes at 45°C. After multiple rinses with universal buffer, streptavidin-horseradish peroxidase detection system (Biomeda, Foster, CA) was applied for 10 minutes. As the final step, the slides were developed for 15 minutes with the enzyme substrate 3 amino-9-ethyl carbazole (AEC, Sigma, St. Louis, MO). The slides were counterstained with hematoxylin solution for 1 minute (Research Genetics, Huntsville, AL). After dehydration, the tissue was sealed with a universal mount (Research Genetics, Huntsville, AL).

Assessments of VEGF and p53 Expression

Assessment of the staining was examined by at least two independent observers without knowledge of the clinical outcomes and a high level of concordance was achieved. Staining intensity of VEGF was graded on a scale with four grades: 0, no staining; 1, weak staining; 2, moderate staining; 3, strong staining, as reported previously1820). The specimens with a score of more than 1 were regarded as positive expression, and those with a score≤1 as negative expression. Immunoreactivity of p53 was graded as follows: positive expression, ≥10% of tumor cells were stained; negative expression, no staining or <10% of tumor cells were stained.

Statistical Analysis

The correlation between VEGF, p53 expression and various clinicopathological parameters was analyzed for statistical significance by the χ2-test and Fisher’s exact probability test. The statistical software program used was Statistical Package for the Social Sciences (SPSS/PC+ 10.0, Chicago, IL). A p-value of less than 0.05 was accepted as statistically significant.

RESULTS

Expression of VEGF and p53 in pancreatic carcinoma

In carcinoma, positive immunostaining for VEGF was identified predominantly in the cytoplasm of the tumor cells (Figure 1). In non-cancerous tissues, islet cells were clearly stained, but acinar and ductal cells were not stained. Positive immunostaining for p53 was present intensely in the nuclei of tumor cells (Figure 2).

Correlation between VEGF and p53 expression

The positive expression of VEGF and p53 was 56.7% (17 out of 30), 60.0% (18 out of 30), respectively. The correlation between VEGF and p53 expression was statistically significant (p=0.004, Table 1).

Correlation between VEGF and p53 expression and clinicopathological parameters

The correlation between VEGF and p53 expression and clinicopathological parameters is summarized in Table 2 and 3 respectively. Positive expression of VEGF correlated with the depth of tumor invasion (p=0.002). There was a trend towards an association between positive expression of VEGF and distant metastasis, although these associations were not statistically significant (p=0.070). VEGF expression did not correlate with patient’s age, sex, tumor size, location, histologic grade, stage or lymph node metastasis. However, no significant correlation was found between p53 expression and various clinicopathological parameters.

DISCUSSION

It has been widely accepted that tumor angiogenesis is one of the most crucial steps in tumor invasion and metastasis35) and many angigenic factors, such as VEGF and basic fibroblast growth factor, have been demonstrated to be involved in such biological behaviors of tumors610). In the current study, we have shown that positive expression of VEGF correlated with the depth of tumor invasion. Also, there was a trend towards an association between positive expression of VEGF and distant metastasis, although these associations were not statistically significant. Leda et al reported that VEGF expression was an independent prognostic factor of the various clinicopathological parameters in pancreatic carcinoma patients23). These results suggest that VEGF, a key factor for the induction of tumor-associated angiogenesis, may be involved in tumor characteristics, including tumor invasion and metastasis. However, Ellis et al reported that VEGF expression was not associated with vessel count and not a predictor of survival or recurrence in pancreatic carcinoma19). Also, Fujimoto et al reported that VEGF expression did not correlate with various clinicopathological parameters, including the depth of tumor invasion, stage, lymph node metastasis or hepatic metastasis20). A discrepancy still exists on the prognostic value of VEGF in pancreatic carcinoma, according to our and other reports. Further studies are warranted to determine the prognostic relevance of VEGF.
Tumor formation and growth are characterized by uncontrolled cellular proliferation. This is usually the result of multiple genetic and epigenetic insults to the cell, particularly involving proto-oncogenes and tumor suppressor genes. The p53 tumor suppressor gene is believed to play a pivotal role in preventing the uncontrolled cellular growth characteristic of a tumor2629). p53 gene is mutated or deleted in about 50% of spontaneously arising tumors2629). Since the cellular half-life of wild type p53 protein is very short and only a very small amount is present in a given cell, normal mucosa is not stained immunohistochemically using monoclonal antibody against this protein. Conversely, the mutated p53 protein accumulates in the nucleus through binding to other oncogenic proteins or by prolonging its half-life28). p53 protein overexpression detected by immunohistochemistry is a prognostic factor in many human neoplasms, such as gastric, colon and pancreatic carcinomas18,38,39).
In our study, the positive expression of p53 was detected in 18 of 30 (60.0%) pancreatic carcinoma tissues analyzed. However, no significant correlation was found between p53 expression and various clinicopathological parameters. There are several possible explanations for this discrepancy. This may in part be due to the relatively small sample size. The protein expression detected by immunohistochemistry is not always in accordance with gene status. Also, the steps of tumor growth and progression are not dependent on mutation of tumor suppressor gene alone and are regulated by many biological processes, including apoptosis, angiogenesis and invasion.
The angiogenic switch is regulated by changes in the relative balance between inducers and inhibitors of endothelial cell proliferation and migration6,7). The switch can be activated by increasing the levels of inducers, such as VEGF and/or by reducing the concentration of inhibitors, such as thrombospondin-16,7). Recently, the status of p53 has been implicated in the regulation of angiogenesis. The presence of wild-type p53 has been associated with the inhibition of angiogenesis, both by increasing expression of thrombospondin-1 and decreasing that of VEGF3034).
Previously, Fujioka et al demonstrated that p53 expression not only was closely related to VEGF expression but also positively correlated with intratumoral microvessel density in pancreatic carcinoma18). Also, our study confirmed a significant correlation between mutant p53 protein and VEGF expression. Thus, p53 mutation during pancreatic tumorigenesis deregulates both arms of the balance, providing a potent stimulus for angiogenesis and tumor progression, although no significant correlation was found between p53 expression and various clinicopathological parameters.
In summary, VEGF, a key factor for the induction of tumor-associated angiogenesis, may be involved in tumor characteristics, including tumor invasion and metastasis. And p53 mutation may be implicated in regulation of angiogenesis through a VEGF up-regulation.

Figure 1.
Immunostaining of vascular endothelial growth factor (VEGF) in pancreatic carcinoma tissues. VEGF expression was identified predominantly in the cytoplasm of the tumor cells (×200).
kjim-17-3-153-1f1.gif
Figure 2.
Immunostaining of p53 in pancreatic carcinoma tissues. Intense nuclear localization of p53 protein was detected in tumor cells (×200).
kjim-17-3-153-1f2.gif
Table 1.
The correlation between VEGF and p53 expression in pancreatic carcinoma
p53 expression VEGF expression
p-value
Negative (n=13) Positive (n=17)
Negative (n=12) 9 3
Positive (n=18) 4 14 0.004
Table 2.
Correlation between VEGF expression and clinicopathological parameters of pancreatic adenocarcinoma
Clinicopathological parameters Total (n=30) Expression of VEGF
p-value
Positive (n=17) Negative (n=13)
Age (years)
  <60 13 7 6 0.785
  ≥60 17 10 7
Sex
  Male 20 11 9 0.794
  Female 10 6 4
Tumor size (cm)
  <4.3 13 6 7 0.310
  ≥4.3 17 11 6
Location
  Head 18 11 7
  Body 7 4 3
  Tail 3 1 2 0.450
  Diffuse 2 1 1
Histologic grade*
  WD 15 10 5 0.552
  MD 7 3 4
  PD 8 4 4
TNM stage
  I 12 8 4
  II 3 2 1
  III 7 5 2 0.212
  IV 8 2 6
Depth of tumor invasion
  T1 4 0 4
  T2 8 4 4
  T3 10 7 3 0.002
  T4 8 6 2
Lymph node metastasis
  Negative 19 11 8 0.858
  Positive 11 6 5
Distant metastasis
  Negative 25 13 12 0.070
  Positive 5 4 1

* WD, well differentiated; MD, moderately differentiated; PD, poorly differentiated adenocarcinoma

Table 3.
Correlation between p53 expression and clinicopathological parameters of pancreatic adenocarcinoma
Clinicopathological parameters Total (n=30) Expression of p53
p-value
Positive (n=18) Negative (n=12)
Age (years)
  <60 13 8 5 0.880
  ≥60 17 10 7
Sex
  Male 20 11 9 0.429
  Female 10 7 3
Tumor size (cm)
  <4.3 13 6 7 0.176
  ≥4.3 17 12 5
Location
  Head 18 10 8
  Body 7 6 1
  Tail 3 2 1 0.162
  Diffuse 2 0 2
Histologic grade*
  WD 15 10 5
  MD 7 3 4 0.561
  PD 8 5 3
TNM stage
  I 12 8 4
  II 3 3 0
  III 7 4 3 0.269
  IV 8 3 5
Depth of tumor invasion
  T1 4 1 3
  T2 8 7 1
  T3 10 7 3 0.083
  T4 8 3 5
Lymph node metastasis
  Negative 19 11 8 0.757
  Positive 11 7 4
Distant metastasis
  Negative 25 15 10 0.963
  Positive 5 3 2

* WD, well differentiated; MD, moderately differentiated; PD, poorly differentiated adenocarcinoma

REFERENCES

1. DiMagno EP, Reber HA, Tempera MA. AGA technical review on the epidemiology, diagnosis and treatment of pancreatic ductal adenocarcinoma. American Gastroenterological Association. Gastroenterolgy 117:1464–14841999.
crossref
2. Livingston EH, Welton ML, Reber HA. Surgical treatment of pancreatic cancer: the United States experience. Int J Pancreatol 9:153–1571991.
crossref pmid pdf
3. Folkman J, Shing Y. Angiogenesis. J Biol Chem 267:10931–109341992.
crossref pmid
4. Folkman J. What is the evidence that tumors are angiogenesis dependent? J Natl Cancer Inst 82:4–61990.
crossref pmid
5. Folkman J. Seminars in Medicine of the Beth Israel Hospital, Boston. Clinical application of research on angiogenesis. N Engl J Med 333:1757–17631995.
crossref pmid
6. Risau W. Mechanisms of angiogenesis. Nature 386:671–6741997.
crossref pmid
7. Hanahan D, Folkman J. Patterns and emerging mechanisms of the angiogenic switch during tumorigenesis. Cell 86:353–3641996.
crossref pmid
8. Leung DW, Cachianes G, Kuang WJ, Goeddel DV, Ferrara N. Vascular endothelial growth factor is a secreted angiogenic mitogen. Science 246:1306–13091989.
crossref pmid
9. Ferrara N, Heinsohn H, Walder CE, Bunting S, Thomas GR. The regulation of blood vessel growth by vascular endothelial growth factor. Ann N Y Acad Sci 752:246–2561995.
crossref pmid
10. Kolch W, Martiny-Baron G, Kieser A, Marme D. Regulation of the expression of the VEGF/VPS and its receptors: role in tumor angiogenesis. Breast Cancer Res Treat 36:139–1551995.
crossref pmid
11. Brown LF, Berse B, Jackman RW, Tognazzi K, Manseau EJ, Dvorak HF, Senger DR. Increased expression of vascular permeability factor (vascular endothelial growth factor) and its receptors in kidney and bladder carcinomas. Am J Pathol 143:1255–12621993.
pmid pmc
12. Brown LF, Berse B, Jackman RW, Tognazzi K, Manseau EJ, Senger DR, Dvorak HF. Expression of vascular permeability factor (vascular endothelial growth factor) and its receptors in adenocarcinomas of the gastrointestinal tract. Cancer Res 53:4727–47351993.
pmid
13. Takahashi Y, Kitadai Y, Bucana CD, Cleary KR, Ellis LM. Expression of vascular endothelial growth factor and its receptor, KDR, correlates with vascularity, metastasis and proliferation of human colon cancer. Cancer Res 55:3964–39681995.
pmid
14. Plate KH, Breier G, Weich HA, Risau W. Vascular endothelial growth factor is a potential tumor angiogenesis factor in human gliomas in vivo. Nature 359:845–8481992.
crossref pmid
15. Kondo S, Asano M, Suzuki H. Significance of vascular endothelial growth, factor/vascular permeability factor for solid tumor growth and its inhibition by the antibody. Biochem Biophys Res Commun 194:1234–12411993.
crossref pmid
16. Kim KJ, Li B, Winer J, Armanini M, Gillett N, Phillips HS, Ferrara N. Inhibition of vascular endothelial growth factor-induced angiogenesis suppresses tumor growth in vivo. Nature 362:841–8441993.
crossref pmid
17. Liu CD, Tilch L, Kwan D, McFadden DW. Vascular endothelial growth factor is increased in ascites from metastatic pancreatic cancer. J Surg Res 102:31–342002.
crossref pmid
18. Fujioka S, Yoshida K, Yanagisawa S, Kawakami M, Aoki T, Yamazaki Y. Angiogenesis in pancreatic carcinoma: thymidine phosphorylase expression in stromal cells and intratumoral microvessel density as independent predictors of overall and relapse-free survival. Cancer 92:1788–17972001.
crossref pmid
19. Ellis LM, Takahashi Y, Fenoglio CJ, Cleary KR, Bucana CD, Evans DB. Vessel counts and vascular endothelial growth factor expression in pancreatic adenocarcinoma. Eur J Cancer 34:337–3401998.
crossref pmid
20. Fujimoto K, Hosotani R, Wada M, Lee JU, Koshiba T, Miyamoto Y, Tsuji S, Nakajima S, Doi R, Imamura M. Expression of two angiogenic factors, vascular endothelial growth factor and platelet-derived endothelial cell growth factor in human pancreatic cancer, and its relationship to angiogenesis. Eur J Cancer 34:1439–14471998.
crossref pmid
21. Itakura J, Ishiwata T, Friess H, Fujii H, Matsumoto Y, Buchler MW, Korc M. Enhanced expression of vascular endothelial growth factor in human pancreatic cancer correlates with local disease progression. Clin Cancer Res 3:1309–13161997.
pmid
22. Baker CH, Solorzano CC, Fidler IJ. Angiogenesis and cancer metastasis: antiangiogenic therapy of human pancreatic adenocarcinoma. Int J Clin Oncol 6:59–652001.
crossref pmid
23. Ikeda N, Adachi M, Taki T, Huang C, Hashida H, Takabayashi A, Sho M, Nakajima Y, Kanehiro H, Hisanaga M, Nakano H, Miyake M. Prognostic significance of angiogenesis in human pancreatic cancer. Br J Cancer 79:1553–15631999.
crossref pmid pmc
24. Arii S, Mori A, Uchida S, Fujimoto K, Shimada Y, Imamura M. Implication of vascular endothelial growth factor in the development and metastasis of human cancers. Hum Cell 12:25–301999.
pmid
25. Seo Y, Baba H, Fukuda T, Takashima M, Sugimachi K. High expression, of vascular endothelial growth factor is associated with liver metastasis and a poor prognosis for patients with ductal pancreatic adenocarcinoma. Cancer 88:2239–22452000.
crossref pmid
26. Hollstein M, Rice K, Greenblatt MS, Soussi T, Fuchs R, Sorlie T, Hovig E, Smith-Sorensen B, Montesano R, Harris CC. Database of p53 gene somatic mutations in human tumors and cell lines. Nucleic Acids Re 22:3551–35551994.

27. Ko LJ, Prives C. p53: puzzle and paradigm. Genes Dev 10:1054–10721996.
crossref pmid
28. Levine AJ. p53, the cellular gatekeeper for growth and division. Cell 88:323–3311997.
crossref pmid
29. Gottlieb TM, Oren M. p53 in growth control and neoplasia. Biochim Biophys Acta 1287:77–1021996.
crossref pmid
30. Dameron KM, Volpert OV, Tainsky MA, Bouck N. Control of angiogenesis in fibroblasts by p53 regulation of thrombospondin-1. Science 265:1582–15841994.
crossref pmid
31. Kieser A, Weich HA, Brandner G, Marme D, Kolch W. Mutant p53 potentiates protein kinase C induction of vascular endothelial growth factor expression. Oncogene 9:963–9691994.
pmid
32. Van Meir EG, Polverini PJ, Chazin VR, Su Huang HJ, de Tribolet N, Cavenee WK. Release of an inhibitor of angiogenesis upon induction of wild-type p53 expression in glioblastoma cells. Nature Genet 8:171–1761994.
crossref pmid
33. Mukhopadhyay D, Tsiokas L, Sukhatme VP. Wild-type p53 and v-Src exert opposing influences on human vascular endothelial growth factor gene expression. Cancer Res 55:6161–61651995.
pmid
34. Bouvet M, Ellis LM, Nishizaki M, Fujiwara T, Liu W, Bucana CD, Fang B, Lee JJ, Roth JA. Adenovirus-mediated wild-type p53 gene transfer down-regulates vascular endothelial growth factor expression and inhibits angiogenesis in human colon cancer. Cancer Res 58:2288–22921998.
pmid
35. In: Fleming ID, Cooper JS, Henson DE, Hutter RVP, Kennedy BJ, Murphy GP, O’Sullivan B, Sobin LH, Yabro JW, eds. American Joint Committee on Cancer, Cancer Staging Manual. Philadelphia: Lippincott-Raven, 121–1261997.

36. Solcia E, Capella C, Kloppel G. Tumors of pancreas. In: Rosai J, Sobin LH, eds. Atlas of Tumor Pathology. third edition. Washington, DC: Armed Forces Institute of Pathology, 31–1441997.

37. Reed JA, Manahan LJ, Park CS, Brigati DJ. Complete one-hour immunohistochemistry based on capillary action. Biotechniques 13:434–4431992.
pmid
38. Kakeji Y, Korenaga D, Tsujitani S, Baba H, Anai H, Maehara Y, Sugimachi K. Gastric cancer with p53 overexpression has high potential for metastasising to lymph nodes. Br J Cancer 67:589–5931993.
crossref pmid pmc
39. Starzynska T, Bromley M, Marlicz K, Roberts SA, Ucunski M, Stern PL. Accumulation of p53 in relation to long-term prognosis in colorectal carcinoma. Eur J Gastroenterol Hepatol 9:183–1861997.
crossref pmid

Editorial Office
101-2501, Lotte Castle President, 109 Mapo-daero, Mapo-gu, Seoul 04146, Korea
Tel: +82-2-2271-6792    Fax: +82-2-790-0993    E-mail: kaim@kams.or.kr                

Copyright © 2024 by Korean Association of Internal Medicine.

Close layer
prev next