Combined Ang-2 and VEGF Targeting Therapies in Renal Cell Carcinoma

Abstract

Angiogenesis, the growth of new vessels from pre-existing ones, is an important feature of tumor growth that has been exploited as a therapeutic target in oncology. Given its key role in facilitating blood vessel sprouting, VEGF has been a major focus of anti-angiogenic strategies, but the observation of resistance in some clinical trials utilizing such agents has led to a search for new or complementary targets in angiogenesis process. The Angiopoietin/Tie2 pathway and in particular the Angiopoietin-2 (Ang-2) ligand which is critically involved in the destabilization of normal vasculature, has been identified as one such target. The current study investigated the potential benefits of combining an Ang-2 targeted therapy with small molecule VEGF targeted agents (Sunitinib, Cediranib) in a human renal cell carcinoma model. The results showed that while both Ang-2 and VEGF interference on their own impaired tumor growth and new blood vessel formation, the combination of agents that targeted both pathways resulted in significantly superior anti-tumor and anti-angiogenic effects.

Share and Cite:

N. Molnar and D. Siemann, "Combined Ang-2 and VEGF Targeting Therapies in Renal Cell Carcinoma," Journal of Cancer Therapy, Vol. 4 No. 9B, 2013, pp. 1-6. doi: 10.4236/jct.2013.49A2001.

Conflicts of Interest

The authors declare no conflicts of interest.

References

[1] M. Papetti and I. M. Herman, “Mechanisms of Normal and Tumor-Derived Angiogenesis,” American Journal of Physiology Cell Physiology, Vol. 282, No. 5, 2002, pp. C947-C970. http://dx.doi.org/10.1152/ ajpcell. 00389.2001
[2] J. Folkman, “Angiogenesis: An Organizing Principle for Drug Discovery?” Nature Reviews Drug Discovery, Vol. 6, No. 4, 2007, pp. 273-286. http://dx.doi.org/10.1038/nrd2115
[3] J. Folkman, “Tumor Angiogenesis: Therapeutic Implications,” New England Journal of Medicine, Vol. 285, No. 21, 1971, pp. 1182-1186. http://dx.doi.org/10.1056/NEJM197111182852108
[4] E. Goldmann, “The Growth of Malignant Disease in Man and the Lower Animals, with Special Reference to the Vascular System,” Proceeding of the Royal Society of Medicine, Vol. 1, Surgery Section, 1908, pp. 1-13.
[5] P. Vaupel, “Abnormal Microvasculature and Defective Microcirculatory Function in Solid Tumors,” In: D. W. Siemann, Ed., Vascular Targeted Therapies in Oncology, Wiley Ltd., West Sussex, 2006, pp. 9-29. http://dx.doi.org/10.1002/0470035439.ch2
[6] J. Denekamp, “Vascular Endothelium as the Vulnerable Element in Tumours,” Acta Radiological Oncology, Vol. 23, No. 4, 1984, pp. 217-225. http://dx.doi.org/10.3109/02841868409136015
[7] K. M. Cook and W. D. Figg, “Angiogenesis Inhibitors: Current Strategies and Future Prospects,” CA: A Cancer Journal for Clinicians, Vol. 60, No. 4, 2010, pp. 222-243. http://dx.doi.org/10.3322/ caac.20075
[8] ClinicalTrials.gov, “Anti-Angiogenic Therapy: Cancer,” U.S. National Institutes of Health, 2013. http://clinicaltrials.gov
[9] C. Coppin, C. Kollmannsberger, L. Le, F. Porzsolt and T. J. Wilt, “Targeted Therapy for Advanced Renal Cell Cancer (RCC): A Cochrane Systematic Review of Published Randomized Trials,” British Journal of Urology International, Vol. 108, No. 10, 2011, pp. 1556-1563.
[10] S. Bellou, G. Pemtheroudakis, C. Murphy and T. Fotsis, “Anti-Angiogenesis in Cancer Therapy: Hercules and Hydra,” Cancer Letters, Vol. 338, No. 2, 2013, pp. 219-228. http://dx.doi.org/10.1016/ j.canlet.2013.05.015
[11] S. Loges, T. Schmidt and P. Carmeliet, “Mechanisms of Resistance to Anti-Angiogenic Therapy and Development of Third-Generation Anti-Angiogenic Drug Candidates,” Genes and Cancer, Vol. 1, No. 1, 2010, pp. 12-25. http://dx.doi.org/10.1177/1947601909356574
[12] G. Bergers and D. Hanahan, “Modes of Resistance to Anti-Angiogenic Therapy,” Nature Reviews Cancer, Vol. 8, No. 8, 2008, pp. 592-603. http://dx.doi.org/10.1038/nrc2442
[13] J. M. Ebos, C. R. Lee and R. S. Kerbel, “Tumor and Host-Mediated Pathways of Resistance and Disease Progression in Response to Antiangiogenic Therapy,” Clinical Cancer Research, Vol. 15, No. 16, 2009, pp. 5020-5025. http://dx.doi.org/10.1158/1078-0432.CCR-09-0095
[14] N. Ferrara, “Pathways Mediating VEGF-Independent Tumor Angiogenesis,” Cytokines & Growth Factor Reviews, Vol. 21, No. 1, 2010, pp. 21-26. http://dx.doi.org/10.1016/j.cytogfr.2009.11.003
[15] D. Gerald, S. Chintharlapalli, H. G. Augustin and L. E. Benjamin, “Angiopoietin-2: An Attractive Target for Improved Antiangiogenic Tumor Therapy,” Cancer Research, Vol. 73, No. 6, 2013. pp. 1649-1657. http://dx.doi.org/10.1158/0008-5472.CAN-12-4697
[16] P. C. Maisonpierre, et al., “Angiopoietin-2, a Natural Antagonist for Tie2 that Disrupts in Vivo Angiogenesis,” Science, Vol. 277, No. 5322, 1997, pp. 55-60. http://dx.doi.org/10.1126/science. 277.5322.55
[17] U. Fiedler, et al., “Angiopoietin-1 and Angiopoietin-2 Share the Same Binding Domains in the Tie-2 Receptor Involving the First Ig-Like Loop and the Epidermal Growth Factor-Like Repeats,” The Journal of Biological Chemistry, Vol. 278, No. 3, 2003, pp. 1721-1727. http://dx.doi.org/10.1074/ jbc.M208550200
[18] W. A. Barton, D. Tzvetkova and D. B. Nikolov, “Structure of the Angiopoietin-2 Receptor Binding Domain and Identification of Surfaces Involved in Tie2 Recognition,” Structure, Vol. 13, No. 5, 2005, pp. 825-832. http://dx.doi.org/10.1016/j.str.2005.03.009
[19] P. Saharinen and K. Alitalo, “The Yin, The Yang, and the Angiopoietin-1,” The Journal of Clinical Investigation, Vol. 121, No. 6, 2011, pp. 2157-2159. http://dx.doi.org/10.1172/JCI58196
[20] M. Yamakawa, et al., “Expression of Angiopoietins in Renal Epithelial and Clear Cell Carcinoma Cells: Regulation by Hypoxia and Participation in Angiogenesis,” American Journal of Physiology Renal Physiology, Vol. 287, No. 4, 2004, pp. F649-F657. http://dx.doi.org/10.1152/ajprenal.00028.2004
[21] M. J. Currie, et al., “Expression of the Angiopoietins and Their Receptor Tie2 in Human Renal Clear Cell Carcinomas; Regulation by the Von Hippel-Lindau Gene and Hypoxia,” Journal of Pathology, Vol. 198, No. 4, 2002, pp. 502-510. http://dx.doi.org/10.1002/path.1228
[22] C. R. Tait and P. F. Jones, “Angiopoietins in Tumours: The Angiogenic Switch,” Journal of Pathology, Vol. 204, No. 1, 2004, pp. 1-10. http://dx.doi.org/10.1002/path.1618
[23] M. Sie, et al., “The Angiopoietin 1/Angiopoietin 2 Balance as a Prognostic Marker in Primary Glioblastoma Multiforme,” Journal of Neurosurgery, Vol. 110, No. 1, 2009, pp. 147-155. http://dx.doi.org/10.3171/ 2008.6.17612
[24] A. J. Lind, et al., “Angiopoietin 2 Expression is Related to Histological Grade, Vascular Density, Metastases, and Outcome in Prostate Cancer,” Prostate, Vol. 62, No. 4, 2005, pp. 394-399. http://dx.doi.org/10.1002/pros.20163
[25] I. Helfrich, et al., “Angiopoietin-2 Levels Are Associated with Disease Progression in Metastatic Malignant Melanoma,” Clinical Cancer Research, Vol. 15, No. 4, 2009, pp. 1384-1392. http://dx.doi.org/10.1158/1078-0432. CCR-08-1615
[26] K. M. Detjen, et al., “Angiopoietin-2 Promotes Disease Progression of Neuroendocrine Tumors,” Clinical Cancer Research, Vol. 16, No. 2, 2010, pp. 420-429. http://dx.doi.org/10.1158/1078-0432.CCR-09-1924
[27] J. H. Park, et al., “Serum Angiopoietin-2 as a Clinical Marker for Lung Cancer,” CHEST, Vol. 132, No. 1, 2007, pp. 200-206. http://dx.doi.org/10.1378/chest.06-2915
[28] G. Bergers and L. E. Benjamin, “Tumorigenesis and the Angiogenic Switch,” Nature Reviews Cancer, Vol. 3, No. 6, 2003, pp. 401-410. http://dx.doi.org/10.1038/nrc1093
[29] H. Hashizume, et al., “Complementary Actions of Inhibitors of Angiopoietin-2 and VEGF on Tumor Angiogenesis and Growth,” Cancer Research, Vol. 70, No. 6, 2010, pp. 2213-2223. http://dx.doi.org/10.1158/ 0008-5472.CAN-09-1977
[30] R. Dandu, et al., “Design and Synthesis of Dihydroindazolo[5,4-A]Pyrrolo[3,4-C]Carbazole Oximes as Potent Dual Inhibitors of TIE-2 and VEGF-R2 Receptor Tyrosine Kinases,” Bioorganic & Medicinal Chemistry Letters, Vol. 18, No. 6, 2008, pp. 1916-1921. http://dx.doi.org/10.1016/ j.bmcl.2008.02.001
[31] Y. J. Koh, et al., “Double Antiangiogenic Protein, DAAP, Targeting VEGF-A and Angiopoietins in Tumor Angiogenesis, Metastasis, and Vascular Leakage,” Cancer Cell, Vol. 18, No. 2, 2010, pp. 171-184. http://dx.doi.org/10.1016/j.ccr.2010.07.001
[32] J. L. Brown, et al., “A Human Monoclonal Anti-ANG2 Antibody Leads to Broad Antitumor Activity in Combination with VEGF Inhibitors and Chemotherapy Agents in Preclinical Models,” Molecular Cancer Therapeutics, Vol. 9, No. 1, 2010, pp. 145-156. http://dx.doi.org/10.1158/1535-7163.MCT-09-0554
[33] C. C. Leow, et al., “MEDI3617, a Human Anti-Angiopoietin 2 Monoclonal Antibody, Inhibits Angiogenesis and Tumor Growth in Human Tumor Xenograft Models,” International Journal of Oncology, Vol. 40, No. 5, 2012, pp. 1321-1330.
[34] V. L. Goodman, et al., “Approval Summary: Sunitinib for the Treatment of Imatinib Refractory or Intolerant Gastrointestinal Stromal Tumors and Advanced Renal Cell Carcinoma,” Clinical Cancer Research, Vol. 13, No. 5, 2007, pp. 1367-1373. http://dx.doi.org/10.1158/1078-0432.CCR-06-2328
[35] A. X. Zhu, et al., “Efficacy, Safety, Pharmacokinetics, and Biomarkers of Cediranib Monotherapy in Advanced Hepatocellular Carcinoma: A Phase II Study,” Clinical Cancer Research, Vol. 19, No. 6, 2013, pp. 1557-1566. http://dx.doi.org/10.1158/1078-0432.CCR-12-3041
[36] R. S. Kerbel, “Antiangiogenic Therapy: A Universal Chemosensitization Strategy for Cancer?” Science, Vol. 312, No. 5777, 2006, pp. 1171-1175. http://dx.doi.org/10.1126/science.1125950

Copyright © 2024 by authors and Scientific Research Publishing Inc.

Creative Commons License

This work and the related PDF file are licensed under a Creative Commons Attribution 4.0 International License.