Effect of tumor microenvironment on tumor VEGF during anti-VEGF treatment: systems biology predictions.
Journal: 2013/August - Journal of the National Cancer Institute
ISSN: 1460-2105
Abstract:
BACKGROUND
Vascular endothelial growth factor (VEGF) is known to be a potent promoter of angiogenesis under both physiological and pathological conditions. Given its role in regulating tumor vascularization, VEGF has been targeted in various cancer treatments, and anti-VEGF therapy has been used clinically for treatment of several types of cancer. Systems biology approaches, particularly computational models, provide insight into the complexity of tumor angiogenesis. These models complement experimental studies and aid in the development of effective therapies targeting angiogenesis.
METHODS
We developed an experiment-based, molecular-detailed compartment model of VEGF kinetics and transport to investigate the distribution of two major VEGF isoforms (VEGF121 and VEGF165) in the body. The model is applied to predict the dynamics of tumor VEGF and, importantly, to gain insight into how tumor VEGF responds to an intravenous injection of an anti-VEGF agent.
RESULTS
The model predicts that free VEGF in the tumor interstitium is seven to 13 times higher than plasma VEGF and is predominantly in the form of VEGF121 (>70%), predictions that are validated by experimental data. The model also predicts that tumor VEGF can increase or decrease with anti-VEGF treatment depending on tumor microenvironment, pointing to the importance of personalized medicine.
CONCLUSIONS
This computational study suggests that the rate of VEGF secretion by tumor cells may serve as a biomarker to predict the patient population that is likely to respond to anti-VEGF treatment. Thus, the model predictions have important clinical relevance and may aid clinicians and clinical researchers seeking interpretation of pharmacokinetic and pharmacodynamic observations and optimization of anti-VEGF therapies.
Relations:
Content
Citations
(34)
References
(84)
Diseases
(1)
Conditions
(1)
Drugs
(1)
Chemicals
(3)
Organisms
(2)
Processes
(1)
Affiliates
(2)
Similar articles
Articles by the same authors
Discussion board
J Natl Cancer Inst 105(11): 802-811

Effect of Tumor Microenvironment on Tumor VEGF During Anti-VEGF Treatment: Systems Biology Predictions

Background

Vascular endothelial growth factor (VEGF) is known to be a potent promoter of angiogenesis under both physiological and pathological conditions. Given its role in regulating tumor vascularization, VEGF has been targeted in various cancer treatments, and anti-VEGF therapy has been used clinically for treatment of several types of cancer. Systems biology approaches, particularly computational models, provide insight into the complexity of tumor angiogenesis. These models complement experimental studies and aid in the development of effective therapies targeting angiogenesis.

Methods

We developed an experiment-based, molecular-detailed compartment model of VEGF kinetics and transport to investigate the distribution of two major VEGF isoforms (VEGF121 and VEGF165) in the body. The model is applied to predict the dynamics of tumor VEGF and, importantly, to gain insight into how tumor VEGF responds to an intravenous injection of an anti-VEGF agent.

Results

The model predicts that free VEGF in the tumor interstitium is seven to 13 times higher than plasma VEGF and is predominantly in the form of VEGF121 (>70%), predictions that are validated by experimental data. The model also predicts that tumor VEGF can increase or decrease with anti-VEGF treatment depending on tumor microenvironment, pointing to the importance of personalized medicine.

Conclusions

This computational study suggests that the rate of VEGF secretion by tumor cells may serve as a biomarker to predict the patient population that is likely to respond to anti-VEGF treatment. Thus, the model predictions have important clinical relevance and may aid clinicians and clinical researchers seeking interpretation of pharmacokinetic and pharmacodynamic observations and optimization of anti-VEGF therapies.

Funding

This work was supported by the National Institutes of Health (R01 CA138264; to ASP and F32 CA154213 to SDF) and the UNCF-Merck Science Initiative (to SDF).

Supplementary Material

Supplementary Data:
Affiliations of authors:Department of Biomedical Engineering (SDF, ASP) and Sidney Kimmel Comprehensive Cancer Center (ASP), Johns Hopkins University School of Medicine, Baltimore MD.
Corresponding author.
Correspondence to: Stacey D. Finley, PhD, Department of Biomedical Engineering, School of Medicine, Johns Hopkins University, 720 Rutland Ave, 613 Traylor Research Bldg, Baltimore, MD 21205 (e-mail ude.uhj@yelnifds).
Received 2012 Jun 28; Revised 2013 Mar 8; Accepted 2013 Mar 22.

Abstract

Background

Vascular endothelial growth factor (VEGF) is known to be a potent promoter of angiogenesis under both physiological and pathological conditions. Given its role in regulating tumor vascularization, VEGF has been targeted in various cancer treatments, and anti-VEGF therapy has been used clinically for treatment of several types of cancer. Systems biology approaches, particularly computational models, provide insight into the complexity of tumor angiogenesis. These models complement experimental studies and aid in the development of effective therapies targeting angiogenesis.

Methods

We developed an experiment-based, molecular-detailed compartment model of VEGF kinetics and transport to investigate the distribution of two major VEGF isoforms (VEGF121 and VEGF165) in the body. The model is applied to predict the dynamics of tumor VEGF and, importantly, to gain insight into how tumor VEGF responds to an intravenous injection of an anti-VEGF agent.

Results

The model predicts that free VEGF in the tumor interstitium is seven to 13 times higher than plasma VEGF and is predominantly in the form of VEGF121 (>70%), predictions that are validated by experimental data. The model also predicts that tumor VEGF can increase or decrease with anti-VEGF treatment depending on tumor microenvironment, pointing to the importance of personalized medicine.

Conclusions

This computational study suggests that the rate of VEGF secretion by tumor cells may serve as a biomarker to predict the patient population that is likely to respond to anti-VEGF treatment. Thus, the model predictions have important clinical relevance and may aid clinicians and clinical researchers seeking interpretation of pharmacokinetic and pharmacodynamic observations and optimization of anti-VEGF therapies.

Abstract

Vascular endothelial growth factor (VEGF) promotes various processes involved in angiogenesis, including endothelial cell proliferation, adhesion, migration, and chemotaxis (1). Angiogenesis is a hallmark of cancer (2) and has been targeted by various cancer therapies, with a focused effort on drugs that inhibit VEGF. Several antiangiogenic agents have been approved by the US Food and Drug Administration (FDA) to treat various cancers and other diseases. Bevacizumab (Genentech, South San Francisco, CA), a recombinant humanized monoclonal antibody to VEGF, is approved for the treatment of metastatic colorectal and kidney cancer, glioblastoma, and non–small cell lung cancer. Ziv-aflibercept (Regeneron, Tarrytown, NY), a soluble decoy receptor for VEGF, is an FDA-approved agent for the treatment of metastatic colorectal cancer and is currently in clinical trials for the treatment of several other cancer types. Other FDA-approved antiangiogenic cancer therapeutics include axitinib, pazopanib, regorafenib, sorafenib, and sunitinib. These agents are small molecule kinase inhibitors with various targets such as VEGF receptors, platelet-derived growth factor receptors, fibroblast growth factor receptors, and Raf kinase.

Systems biology approaches are useful in gaining a broader understanding of the complexity of angiogenesis. Computational models can be applied to generate and test biological hypotheses and can aid in the development of effective therapies that target angiogenesis (3). Additionally, models can provide a framework to predict promising drug targets and identify patient populations that will respond to a particular therapy.

We have developed a molecular-detailed compartment model that is useful in understanding VEGF dynamics in the body. The model is based on detailed biochemical kinetics and molecular transport and has been validated against available experimental data. It is a predictive tool that can provide insight into the distribution of VEGF in the body and the effects of systemic administration of anti-VEGF therapeutics, such as bevacizumab and aflibercept. We have applied the model to understand and explain clinical observations of anti-VEGF agents (4) and predict the effect of the drugs (5,6). Here, we present three important model predictions regarding the pretreatment levels of VEGF121 and VEGF165 and the dynamic response of plasma and tumor VEGF to anti-VEGF treatment. We compare our results with available experimental data and propose clinical applications of the model predictions.

* See Supplementary Table 1 (available online).

† No data available.

Click here to view.

Notes

SDF and ASP designed the study. SDF performed the simulations and wrote the manuscript. All authors analyzed results and prepared the final manuscript.

We thank Drs Feilim Mac Gabhann and Herbert I. Hurwitz for their constructive comments and members of our laboratory for helpful discussions.

Notes

References

  • 1. Jayson GC, Hicklin DJ, Ellis LM. Antiangiogenic therapy—evolving view based on clinical trial results. Nat Rev Clin Oncol. 2012;9(5):297–303 [[PubMed]
  • 2. Hanahan D, Wienberg RA. Hallmarks of cancer: the next generation. Cell. 2011;144(5):646–674 [[PubMed]
  • 3. Arrell DK, Terzic A. Network systems biology for drug discovery. Clin Pharmacol Ther. 2010;8(1):120–125 [[PubMed]
  • 4. Stefanini MO, Wu FTH, Mac Gabhann F, Popel AS. Increase of plasma VEGF after intravenous administration of bevacizumab is predicted by a pharmacokinetic model. Cancer Res. 2010;70(23):9886–9894
  • 5. Finley SD, Popel AS. Predicting the effects of anti-angiogenic agents targeting specific VEGF isoforms. AAPS J. 2012;14(3):500–509
  • 6. Finley SD, Engel-Stefanini MO, Imoukhuede PI, Popel AS. Pharmacokinetics and pharmacodynamics of VEGF-neutralizing antibodies. BMC Syst Biol. 2011;5:193
  • 7. Gavin TP, Ruster RS, Carrithers JA, et al No difference in the skeletal muscle angiogenic response to aerobic exercise training between young and aged men. J Physiol. 2007;585(1):231–239 [Google Scholar]
  • 8. Rullman E, Rundqvist H, Wagsater D, et al A single bout of exercise activates matrix metalloproteinase in human skeletal muscle. J Appl Physiol. 2007;102(6):2346–2351 [[PubMed][Google Scholar]
  • 9. Hellsten Y, Rufener N, Nielsen JJ, Hoier B, Krustrup P, Bangsbo J. Passive leg movement enhances interstitial VEGF protein, endothelial cell proliferation, and eNOS mRNA content in human skeletal muscle. Am J Physiol Regul Integr Comp Physiol. 2008;294(3):R975–R982 [[PubMed]
  • 10. Hansen-Algenstaedt N, Nielsen JJ, Saltin B, Hellsten Y. Exercise training normalizes skeletal muscle vascular endothelial growth factor levels in patients with essential hypertension. J Hypertens. 2010;28(6): 1176–1185 [[PubMed]
  • 11. Hoier B, Rufener N, Bojsen-Moller J, Bangsbo J, Hellsten Y. The effect of passive movement training on angiogenic factors and capillary growth in human skeletal muscle. J Physiol. 2010;588(19):3833–3845
  • 12. Hoier B, Nordsborg N, Andersen S, et al Pro- and anti-angiogenic factors in human skeletal muscle in response to acute exercise and training. J Physiol. 2012;590(3):595–606 [Google Scholar]
  • 13. Hoier B, Passos M, Bangsbo J, Hellsten Y. Intense intermittent exercise provides weak stimulus for VEGF secretion and capillary growth in skeletal muscle. Exp Physiol. 2013;98(2):585–597 [[PubMed]
  • 14. Hoar FJ, Lip GYH, Belgore F, Stonelake PS. Circulating levels of VEGF-A, VEGF-D and soluble VEGF-A receptor (sFlt-1) in human breast cancer. Int J Biol Markers. 2004;19(3):229–235 [[PubMed]
  • 15. Kut C, Mac Gabhann F, Popel AS. Where is VEGF in the body? A meta-analysis of VEGF distribution in cancer. Br J Cancer. 2007;97(7):978–985
  • 16. Aref S, El Sherbiny M, Goda T, Fouda M, Al Askalany H, Abdalla D. Soluble VEGF/sFLt1 ratio is an independent predictor of AML patient out come. Hematology. 2005;10(2):131–134 [[PubMed]
  • 17. Caine GJ, Blann AD, Stonelake PS, Ryan P, Lip GYH. Plasma angiopoietin-2 ant tie-2 in breast and prostate cancer: a comparison with VEGF and flt-1. Eur J Clin Invest. 2003;33(10):883–890 [[PubMed]
  • 18. Ustuner A, Saip P, Yasasever V, et al Prognostic and predictive value of vascular endothelial growth factor and its soluble receptors, VEGFR-1 and VEGFR-2 levels in the sera of small cell lung cancer patients. Med Oncol. 2008;25(4):394–399 [[PubMed][Google Scholar]
  • 19. Chang YT, Chang MC, Wei SC, et al Serum vascular endothelial growth factor/soluble vascular endothelial growth factor receptor 1 ratio is an independent prognostic marker in pancreatic cancer. Pancreas. 2008;37(2):145–150 [[PubMed][Google Scholar]
  • 20. Chauhan VP, Stylianopoulos T, Martin JD, et al Normalization of tumour blood vessels improves the delivery of nanomedicines in a size-dependent manner. Nat Nanotechnol. 2012;7(6):383–388 [Google Scholar]
  • 21. Duysinx BC, Corhay J-L, Hubin L, Nguyen D, Henket M, Louis R. Diagnostic value of interleukin-6, transforming growth factor-beta 1 and vascular endothelial growth factor in malignant pleural effusions. Respir Med. 2008;102(12):1708–1714 [[PubMed]
  • 22. Economidou F, Antoniou KM, Tzanakis N, Sfiridaki K, Siafakas NM, Schiza SE. Angiogenic molecule Tie-2 and VEGF in the pathogenesis of pleural effusions. Respir Med. 2008;102(5):774–779 [[PubMed]
  • 23. Gieseler F, Luhr I, Kunze T, et al Activated coagulation factors in human malignant effusions and their contribution to cancer cell metastasis and therapy. Thromb Haemost. 2007;97(6):1023–1030 [[PubMed][Google Scholar]
  • 24. Hamed EA, El-Noweihi AM, Mohamed AZ, Mahmoud A. Vasoactive mediators (VEGF and TNF-a) in patients with malignant and tuberculous pleural effusions. Respirology. 2004;9(1):81–86 [[PubMed]
  • 25. Harlozinska A, Sedlaczek P, Kulpa J, et al Vascular endothelial growth factor (VEGF) concentration in sera and tumor effusions from patients with ovarian carcinoma. Anticancer Res. 2004;24(2C):1149–1158 [[PubMed][Google Scholar]
  • 26. Ishimoto O, Saijo Y, Narumi K, et al High level of vascular endothelial growth factor in hemorrhagic pleural effusion of cancer. Oncology. 2002;63(1):70–75 [[PubMed][Google Scholar]
  • 27. Jin HY, Lee KS, Jin SM, Lee Y-C. Vascular endothelial growth factor correlates with matrix metalloproteinase-9 in the pleural effusion. Respir Med. 2004;98(2):115–122 [[PubMed]
  • 28. Kishiro I, Kato S, Fuse D, Yoshida T, Machida S, Kaneko N. Clinical significance of vascular endothelial growth factor in patients with primary lung cancer. Respirology. 2002;7(2):93–98 [[PubMed]
  • 29. Kotyza J, Havel D, Vrzalova J, Kulda V, Pesek M. Diagnostic and prognostic significance of inflammatory markers in lung cancer-associated pleural effusions. Int J Biol Markers. 2010;25(1):12–20 [[PubMed]
  • 30. Kraft A, Weindel K, Ochs A, et al Vascular endothelial growth factor in the sera and effusions of patients with malignant and nonmalignant disease. Cancer Metastasis Rev. 1999;85(1):178–187 [[PubMed][Google Scholar]
  • 31. Lee HK, Chae HS, Kim JS, et al Vascular endothelial growth factor levels in ascites between chemonaive and chemotreated patients. Yonsei Med J. 2008;49(3):429–435 [Google Scholar]
  • 32. Matsuyama W, Hashiguchi T, Mizoguchi A, et al Serum levels of vascular endothelial growth factor dependent on the stage progression of lung cancer. Chest. 2000;118(4):948–951 [[PubMed][Google Scholar]
  • 33. Nascimento I, Schaer R, Lemaire D, et al Vascular endothelial growth factor (VEGF) levels as a tool to discriminate between malignant and nonmalignant ascites. Acta Pathol Microbiol Scand. 2004;112(9):585–587 [[PubMed][Google Scholar]
  • 34. Richardson M, Gunawan J, Hatton MWC, Seidlitz E, Hirte HW, Signh G. Malignant ascites fluid (MAF), including ovarian-cancer-associated MAF, contains angiogstatin and other factor(s) which inhibit angiogenesis. Gynecol Oncol. 2002;86(3):279–287 [[PubMed]
  • 35. Rudlowski C, Pickarts A-K, Fuhljahns C, et al Prognostic significance of vascular endothelial growth factor expression in ovarian cancer patients: a long-term follow-up. Int J Gynecol Cancer. 2006;16(S1):183–189 [[PubMed][Google Scholar]
  • 36. Sack U, Hoffmann M, Zhao XJ, et al Vascular endothelial growth factor in pleural effusions of different origin. Eur Respir J. 2005;25(4):600–604 [[PubMed][Google Scholar]
  • 37. Shu J, Sun G, Liu H, Liu J. Clinical utility of vascular endothelial growth factor in diagnosing malignant pleural effusions. Acta Oncol. 2007;46(7): 1004–1011 [[PubMed]
  • 38. Sun X-M, Dont W-G, Gao L-C. Detection of VEGF levels in ascites and peritoneal fluid. Chin J Cancer Res. 2003;15(4):310–314 [PubMed]
  • 39. Thickett DR, Armstrong L, Millar AB. Vascular endothelial growth factor (VEGF) in inflammatory and malignant pleural effusions. Thorax. 1999;54(8):707–710
  • 40. Verheul HM, Hoekman K, Jorna AS, Smit EF, Pinedo HM. Targeting vascular endothelial growth factor blockade: ascites and pleural effusion formation. Oncologist. 2000;5(Suppl 1):45–50 [[PubMed]
  • 41. Xue K, Siong S, Xiong W. Clincial value of vascular endothelial growth factor combined with interferon-gamma in diagnosing malignant pleural effusion and tuberculous pleural effusion. J Huazhong Univ Sci Technol (Med Sci). 2007;27(5):495–497 [[PubMed]
  • 42. Yabushita H, Shimazu M, Noguchi M, et al Vascular endothelial growth factor activating matrix metalloproteinase in ascitic fluid during peritoneal dissemination of ovarian cancer. Oncol Rep. 2003;10(1):89–95 [[PubMed][Google Scholar]
  • 43. Yamamoto S, Konishi I, Mandai M, et al Expression of vascular endothelial growth factor (VEGF) in epithelial ovarian neoplasms: correlation with clinicopathology and patient survival, and analysis of serum VEGF levels. Br J Cancer. 1997;76(9):1221–1227 [Google Scholar]
  • 44. Yanagawa H, Takeuchi E, Suzuki Y, Ohmoto Y, Bando H, Sone S. Vascular endothelial growth factor in malignant pleural effusion associated with lung cancer. Cancer Immunol Immunother. 1999;48(7):396–400 [[PubMed]
  • 45. Zebrowski BK, Yano S, Liu W, et al Vascular endothelial growth factor levels and induction of permeability in malignant pleural effusions. Clin Cancer Res. 1999;5(11):3364–3368 [[PubMed][Google Scholar]
  • 46. Zhou W-B, Bai M, Jin Y. Diagnostic value of vascular endothelial growth factor and endostatin in malignant pleural effusions. Int J Tuberc Lung Dis. 2009;13(3):381–386 [[PubMed]
  • 47. Hoskins ER, Hood BL, Sun M, Krivak TC, Edwards RP, Conrads TP. Proteomic analysis of ovarian cancer proximal fluids: validation of elevated peroxiredoxin 1 in patient perifpheral circulation. PLoS One. 2011;6(9):e25056
  • 48. Garvin S, Dabrosin C. In vivo measurement of tumor estradiol and vascular endothelial growth factor in breast cancer patients. BMC Cancer. 2008;8:73
  • 49. Wiig H, Tenstad O, Iversen PO, Kalluri R, Bjerkvig R. Interstitial fluid: the overlooked component of the tumor microenvironment?Fibrogenesis and Tissue Repair. 2010;3:12
  • 50. Ljungberg B, Jacobsen J, Haggstrom-Rudolfssson S, Rasmuson T, Lindh G, Grankvist K. Tumor vascular endothelial growth factor (VEGF) mRNA in relation to serum VEGF protein levels and tumour progression in human renal cell carcinoma. Urol Res. 2003;31(5):335–340 [[PubMed]
  • 51. Stimpfl M, Tong D, Fasching B, et al Vascular endothelial growth factor splice variants and their prognostic value in breast and ovarian cancer. Clin Cancer Res. 2002;8(7):2253–2259 [[PubMed][Google Scholar]
  • 52. Yuan A, Yu CJ, Luh KT, Lin FY, Kuo SH, Yang PC. Quantification of VEGF mRNA expression in non-small cell lung cancer using a real-time quantitative reverse transcription-PCR assay and a comparison with quantitative competitive reverse transcription-PCR. Lab Invest. 2000;2000(80):11 [[PubMed]
  • 53. Zygalaki E, Tsaroucha EG, Kaklamanis L, Lianidou ES. Quantitative real-time reverse transcription-PCR study of the expression of vascular endothelial growth factor (VEGF) splice variants and VEGF receptors (VEGFR-1 and VEGFR-2) in non-small cell lung cancer. Clin Chem. 2007;53(8):1433–1439 [[PubMed]
  • 54. Ng YS, Rohan R, Sunday ME, Demello DE, D’Amore PA. Differential expression of VEGF isoforms in mouse during development and in the adult. Dev Dyn. 2001;220(2):112–121 [[PubMed]
  • 55. Houck K, Leung DW, Rowland AM, Winer J, Ferrara N. Dual regulation of vascular endothelial growth factor bioavailability by genetic and proteolytic mechanisms. J Biol Chem. 1992;268(36):26031–26037 [[PubMed]
  • 56. Soker S, Miao H-Q, Nomi M, Takashima S, Klagsbrun M. VEGF165 mediates formation of complexes containing VEGFR-2 and neuropilin-1 that enhance VEGF165-receptor binding. J Cell Biochem. 2002;85(2):357–368 [[PubMed]
  • 57. Poon RT-P, Lau CP-Y, Cheung S-T, Yu W-C, Fan S-T. Quantitative correlation of serum levels and tumor expression of vascular endothelial growth factor in patients with hepatocellular carcinoma. Cancer Res. 2003;63(12):3121–3216 [[PubMed]
  • 58. Poon RT-P, Lau CP-Y, Ho JW-Y, Yu W-C, Fan S-T, Wong J. Tissue factor expression correlates with tumor angiogenesis and invasiveness in human hepatocellular carcinoma. Clin Cancer Res. 2003;9(14):5339–5345 [[PubMed]
  • 59. Gutierrez J, Konecny GE, Hong K, et al A new ELISA for use in a 3-ELISA system to assess concentrations of VEGF splice variants and VEGF110 in ovarian cancer tumors. Clin Chem. 2008;54(3):597–601 [[PubMed][Google Scholar]
  • 60. Segerstrom L, Fuchs D, Backman U, Holmquist K, Christofferson R, Azarbayjani F. The anti-VEGF antibody bevacizumab potently reduces the growth rate of high-risk neuroblastoma xenografts. Pediatr Res. 2006; 60(5):576–581 [[PubMed]
  • 61. Willett CG, Boucher Y, Duda DG, et al Surrogate markers for antiangiogenic therapy and dose-limiting toxicities for bevacizumab with radiation and chemotherapy: continued experience of a phase I trial in rectal cancer patients. J Clin Oncol. 2005;23(31):8136–8139 [[PubMed][Google Scholar]
  • 62. Yang JC, Haworth L, Sherry RM, et al A randomized trial of bevacizumab, an anti-vascular endothelial growth factor antibody, for metastatic renal cancer. New Engl J Med. 2003;349(5):427–434 [Google Scholar]
  • 63. Lee J, Ku T, Yu H, et al Blockade of VEGF-A suppresses tumor growth via inhibition of autocrine signaling through FAK and AKT. Cancer Lett. 2012;318(2):221–225 [[PubMed][Google Scholar]
  • 64. Cao Y, E G, Wang E, et al VEGF exerts an angiogenesis-independent function in cancer cells to promote their malignant progression. Cancer Res. 2012;72(16):3912–3918 [Google Scholar]
  • 65. Adamcic U, Skowronski K, Peters C, Morrison J, Coomber BL. The effect of bevacizumab on human malignant melanoma cells with function VEGF/VEGFR2 autocrine and intracrine signaling loops. Neoplasia. 2012;14(7):612–623
  • 66. Lee TH, Seng S, Sekine M, Hinton C, Avraham HK, Avraham S. Vascular endothelial growth factor mediates intracrine survival in human breast carcinoma cells through internally expressed VEGFR1/FLT1. PLoS Med. 2007;4(6):e186
  • 67. Zhang J, Jia Z, Li Q, et al Elevated expression of vascular endothelial growth factor correlates with increased angiogenesis and decreased progression-free survival among patients with low-grade neuroendocrine tumors. Cancer. 2007;109(8):1478–1486 [[PubMed][Google Scholar]
  • 68. Ricci AG, Olivares CN, Biolotas MA, Meresman GF, Baranao RI. Effect of vascular endothelial growth factor inhibition on endometrial implant development in a murine model of endometriosis. Reprod Sci. 2011;18(7):614–622 [[PubMed]
  • 69. Lan K-L, Ou-Yang F, Yen S-H, Shih H-L, Lan K-H. Cationic liposome coupled endostatin gene for treatment of peritoneal colon cancer. Clin Exp Metastasis. 2010;27(5):307–318 [[PubMed]
  • 70. Okada Y, Akisue T, Hara H, et al The effect of bevacizumab on tumour growth of malignant fibrous histiocytoma in an animal model. Anticancer Res. 2010;30(9):3391–3395 [[PubMed][Google Scholar]
  • 71. Yuan A, Lin C-Y, Chou C-H, et al Functional and structural characteristics of tumor angiogenesis in lung cancers overexpressing different VEGF isoforms assessed by DCE- and SSCE-MRI. PLoS One. 2011;6(1):e16062 [Google Scholar]
  • 72. Rennel ES, Harper SJ, Bates DO. Therapeutic potential of manipulating VEGF splice isoforms in oncology. Future Oncol. 2009;5(5):703–712
  • 73. Dokun AO, Annex BH. The VEGF165b “ICE-o-form” puts a chill on the VEGF story. Circ Res. 2011;109(3):246–247
  • 74. Manetti M, Guiducci S, Romano E, et al Overexpression of VEGF165b, an inhibitory splice variant of vascular endothelial growth factor, leads to insufficient angiogenesis in patients with systemic sclerosis. Circ Res. 2011;109(3):e14–e26 [[PubMed][Google Scholar]
  • 75. Nowak DG, Woolard J, Amin EM, et al Expression of pro- and anti-angiogenic isoforms of VEGF is differentially regulated by splicing and growth factors. J Cell Sci. 2008;121(20):3487–3495 [Google Scholar]
  • 76. Catena R, Larzabal L, Larrayoz M, et al VEGF121b and VEGF165b are weakly angiogenic isoforms of VEGF-A. Mol Cancer. 2010;9:320 [Google Scholar]
  • 77. Italiano JE, Richardson JL, Patel-Hett S, et al Angiogenesis is regulated by a novel mechanism: pro- and antiangiogenic proteins are organized into separate platelet a-granules and differentially released. Blood. 2008;111(3): 1227–1233 [Google Scholar]
  • 78. Battinelli EM, Markens BA, Italiano JE., Jr Release of angiogenesis regulatory proteins from platelet alpha granules: modulation of physiologic and pathologic angiogenesis. Blood. 2011;118(5):1359–1369
  • 79. Verheul HMW, Lolkema MPJ, Quian DZ, et al Platelets take up the monoclonal antibody bevacizumab. Clin Cancer Res. 2007;13(18):5341–5347 [[PubMed][Google Scholar]
  • 80. Zhang Q-X, Magover CJ, Mack CA, Budenbender KT, Ko W, Rosengart T. Vascular endothelial growth factor is the major angiogenic factor in omentum: mechanism of the omentum-mediated angiogenesis. J Surg Res. 1997;67(2):147–154 [[PubMed]
  • 81. Maharaj ASR, D’Amore PA. Roles for VEGF in adult. Microvasc Res. 2007;74:100–113
  • 82. Grunstein J, Masbad JJ, Hickey R, Giordano F, Johnson RS. Isoforms of vascular endothelial growth factor act in a coordinate fashion to recruit and expand tumor vasculature. Mol Cell Biol. 2000;20(19):7292–7291
  • 83. Tozer GM, Akerman S, Cross NA, et al Blood vessel maturation and response to vascular-disrupting therapy in single vascular endothelial growth factor-A isoform-producing tumors. Cancer Res. 2008;68(7):2301–2311 [[PubMed][Google Scholar]
  • 84. Sanmartin E, Jantus E, Blasco A, et al Plasma levels of VEGF-A and VEGFR-2 in advanced NSCLC. J Clin Oncol. 2010;28(15s):10623 [PubMed][Google Scholar]
  • 85. Bernaards C, Hegde P, Chen D, et al Circulating vascular endothelial growth factor (VEGF) as a biomarker for bevacizumab-based therapy in metastatic colorectal, non-small cell lung, and renal cell cancers: analysis of phase III studies. J Clin Oncol. 2010;28(15s):10519 [PubMed][Google Scholar]
  • 86. Jubb AM, Miller KD, Rugo HS, et al Impact of exploratory biomarkers on the treatment effect of bevacizumab in metastatic breast cancer. Clin Cancer Res. 2011;17(2):372–381 [Google Scholar]
  • 87. Van Cutsem E, de Haas S, Kang Y-K, et al Bevacizumab in combination with chemotherapy as first-line therapy in advanced gastric cancer: a biomarker evaluation from the AVAGAST randomized phase III trial. J Clin Oncol. 2012;30(17):2119–2127 [[PubMed][Google Scholar]
Collaboration tool especially designed for Life Science professionals.Drag-and-drop any entity to your messages.