Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Review Article
  • Published:

Angiogenesis modulation in cancer research: novel clinical approaches

Key Points

  • For a tumour to grow beyond a certain size, it must develop a network of blood vessels to supply nutrients and oxygen and to remove waste products.

  • Advances in the understanding of this process of tumour angiogenesis have led to the development of many drugs that target the different steps that are involved, more than 30 of which have entered clinical trials. However, so far, none has been approved, and there have been several prominent failures, which might be because clinical approaches that were established for traditional cytotoxic agents are inappropriate for angiogenic modulators.

  • The usual clinical development of a cytotoxic agent is based on the following concepts: first, that the agent is associated with dose-dependent toxicity; second, that there is an upper limit for dose escalation, which is defined as the dose-limiting toxicity (DLT); third, that the maximum-tolerated dose (MTD) has a higher probability of shrinking tumours (defined as objective remission) and improving palliation of symptoms; and finally, that the agent or combination regimens that are associated with tumour shrinkage might prolong survival.

  • By contrast, in early Phase I/II trials, angiogenic modulators have shown modest toxic effects and are mainly cytostatic, slowing or stopping the tumour growth and the development of metastases without producing an objective remission.

  • It seems clear that the end points for dose-defining trials (Phase I) and efficacy trials (Phase II) should be reconsidered. We strongly recommend the extensive use of correlative studies in the early phases of drug development to establish surrogate biomarkers for use in efficacy trials. Novel Phase II trial designs should be considered to address issues such as the low probability of an objective measurable response with angiogenic modulators.

  • Imaging studies could have a key role in assessing the efficacy of treatments. Various imaging modalities, such as magnetic resonance imaging, ultrasonography, positron emission tomography and computed tomography, can be selected for this purpose; the choice of imaging studies should be based on an accurate evaluation of the novel agents in preclinical models.

  • Finally, careful selection of the clinical setting for the investigation (for example, tumour type and stage of disease) must be carried out before expensive, definitive Phase III clinical trials are initiated.

Abstract

Angiogenesis — the formation of new blood vessels — is essential for tumour progression and metastasis. Consequently, the modulation of tumour angiogenesis using novel agents has become a highly active area of investigation in cancer research, from the bench to the clinic. However, the great therapeutic potential of these agents has yet to be realized, which could, in part, be because the traditional strategies that are used in clinical trials for anticancer therapies are not appropriate for assessing the efficacy of agents that modulate angiogenesis. Here, we discuss methods for monitoring the biological activity of angiogenic modulators, and innovative approaches to trial design that might facilitate the integration of these agents into anticancer therapy.

This is a preview of subscription content, access via your institution

Access options

Buy this article

USD 39.95

Prices may be subject to local taxes which are calculated during checkout

Figure 1: Simplified overview of some key steps in tumour angiogenesis.
Figure 2: Representation of the clinical drug development process.
Figure 3: Imaging studies to monitor tumour angiogenesis.

Similar content being viewed by others

References

  1. Carmeliet, P. Mechanisms of angiogenesis and arteriogenesis. Nature Med. 6, 389–395 (2000).

    Article  CAS  PubMed  Google Scholar 

  2. Folkman, J. Tumour angiogenesis: therapeutic implications. N. Engl. J. Med. 285, 1182–1186 (1971).

    Article  CAS  PubMed  Google Scholar 

  3. Skobe, M. et al. Halting angiogenesis suppresses carcinoma cell invasion. Nature Med. 3, 1222–1227 (1997).

    Article  CAS  PubMed  Google Scholar 

  4. Zhang, H. T. et al. Enhancement of tumour growth and vascular density by transfection of vascular endothelial cell growth factor into MCF-7 human breast carcinoma cells. J. Natl Cancer Inst. 87, 213–219 (1995).

    Article  CAS  PubMed  Google Scholar 

  5. Carmeliet, P. & Jain, R. K. Angiogenesis in cancer and other diseases. Nature 407, 249–257 (2001).

    Article  Google Scholar 

  6. Yancopoulos, G. D. et al. Vascular-specific growth factors and blood vessel formation. Nature 407, 242–248 (2000).

    Article  CAS  PubMed  Google Scholar 

  7. Hanahan, D. & Weinberg, R. A. The hallmarks of cancer. Cell 100, 57–70 (2000).

    CAS  PubMed  Google Scholar 

  8. Bouck, N., Stellmach, V. & Hsu, S. C. How tumours become angiogenic. Adv. Cancer Res. 69, 135–174 (1996).

    Article  CAS  PubMed  Google Scholar 

  9. Robinson, C. J. et al. The splice variants of vascular endothelial growth factor (VEGF) and their receptors. J. Cell Sci. 114, 853–865 (2001).

    CAS  PubMed  Google Scholar 

  10. Hiraoka, N. et al. Matrix metalloproteinases regulate neovascularization by acting as pericellular fibrinolysins. Cell 91, 365–377 (1998).

    Article  Google Scholar 

  11. Levine, H. A. et al. A mathematical model for the roles of pericytes and macrophages in the initiation of angiogenesis. The role of protease inhibitors in preventing angiogenesis. Mathem. Biosc. 168, 77–115 (2000).

    Article  CAS  Google Scholar 

  12. Sturzebecher, J. et al. 3-Amidinophenylalanine-based inhibitors of urokinase. Bioorg. Med. Chem. Lett. 9, 3147–3152 (1999).

    Article  CAS  PubMed  Google Scholar 

  13. Presta, L. G. et al. Humanization of an anti-vascular endothelial growth factor monoclonal antibody for the therapy of solid tumours and other disorders. Cancer Res. 57, 4593–4599 (1997).This paper is an extremely interesting example of the progress of an angiogenic modulator in more advanced clinical development.

    CAS  PubMed  Google Scholar 

  14. Shaheen, R. M. et al. Antiangiogenic therapy targeting the tyrosine kinase receptor for vascular endothelial growth factor receptor inhibits the growth of colon cancer liver metastasis and induces tumour and endothelial cell apoptosis. Cancer Res. 59, 5412–5416 (1999).

    CAS  PubMed  Google Scholar 

  15. Oshika, Y. et al. Ribozyme approach to downregulate vascular endothelial growth factor (VEGF) 189 expression in non-small cell lung cancer (NSCLC). Eur. J. Cancer 36, 2390–2396 (2000).

    Article  CAS  PubMed  Google Scholar 

  16. Westphal, J. R. et al. Angiostatin generation by human tumour cell lines: involvement of plasminogen activators. Int. J. Cancer 86, 760–767 (2000).

    Article  CAS  PubMed  Google Scholar 

  17. Huang, X. et al. Soluble recombinant endostatin purified from Escherichia coli: antiangiogenic activity and antitumour effect. Cancer Res. 61, 478–481 (2001).

    CAS  PubMed  Google Scholar 

  18. Fotsis, T. et al. The endogenous oestrogen metabolite 2-methoxyestradiol inhibits angiogenesis and suppresses tumour growth. Nature 368, 237–239 (1994).

    Article  CAS  PubMed  Google Scholar 

  19. Pasqualini, R. et al. Aminopeptidase N is a receptor for tumour-homing peptides and a target for inhibiting angiogenesis. Cancer Res. 60, 722–727 (2000).

    CAS  PubMed  PubMed Central  Google Scholar 

  20. Chen, J., Baskerville, C., Han, Q., Pan, Z. K., & Huang, S. αv-Integrin, p38 mitogen-activated protein kinase, and urokinase plasminogen activator are functionally linked in invasive breast cancer cells. J. Biol. Chem. 276, 47901–47905 (2001).

    Article  CAS  PubMed  Google Scholar 

  21. O-Charoenrat, P., Rhys-Evans, P., Modjtahedi, H. & Eccles, S. A. Vascular endothelial growth factor family members are differentially regulated by c-erbB signaling in head and neck squamous carcinoma cells. Clin. Exp. Metastasis 18, 155–161 (2000).

    Article  CAS  PubMed  Google Scholar 

  22. Ciardiello, F. et al. Inhibition of growth factor production and angiogenesis in human cancer cells by ZD1839 (Iressa), a selective epidermal growth factor receptor tyrosine kinase inhibitor. Clin. Cancer Res. 7, 1459–1465 (2001).

    CAS  PubMed  Google Scholar 

  23. Rak, J. et al. Ras regulation of vascular endothelial growth factor and angiogenesis. Methods Enzymol. 333, 267–283 (2001).

    Article  CAS  PubMed  Google Scholar 

  24. Ang, Y. et al. Aberrant p53 expression correlates with expression of vascular endothelial growth factor mRNA and interleukin-8 mRNA and neoangiogenesis in non-small cell lung cancer. J. Clin. Oncol. 20, 900–910 (2002).

    Google Scholar 

  25. Giatromanolaki, A. et al. Relation of hypoxia inducible factor 1α and 2α in operable non-small cell lung cancer to angiogenic/molecular profile of tumours and survival. Br. J. Cancer 85, 881–890 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Sunwoo, J. B. et al. Novel proteasome inhibitor PS-341 inhibits activation of nuclear factor-κB, cell survival, tumour growth, and angiogenesis in squamous cell carcinoma. Clin. Cancer Res. 7, 1419–1429 (2001).

    CAS  PubMed  Google Scholar 

  27. Okada, A. et al. Membrane-type matrix metalloproteinase (MT-MMP) gene is expressed in stromal cells of human colon, breast and head and neck carcinoma. Proc. Natl Acad. Sci. USA 92, 2730–2734 (1995).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Dewhrist, M. W. et al. Morphologic and hemodynamic comparison of tumour and healing normal tissue microvasculature. Int. J. Radiat. Oncol. Biol. Phys. 17, 91–99 (1989).

    Article  Google Scholar 

  29. Lal, B. K., Varma, S., Pappas, P. J., Hobson, R. W. & Duran, W. N. VEGF increases permeability of the endothelial cell by activation of PKB/Akt, endothelial cell nitric-oxide synthase, and MAP kinase pathways. Microvasc. Res. 62, 252–262 (2001).

    Article  CAS  PubMed  Google Scholar 

  30. Helmlinger, G., Yuan, F., Dellian, M. & Jain, R. K. Interstitial pH and pO2 gradients in solid tumours in vivo: simultaneous high-resolution measurements reveal a lack of correlation. Nature Med. 3, 177–182 (1997).An extremely accurate description of the modifications in the 'tumour microenviroment'. It will stimulate interest in reading and searching further information on this topic that could apply to diagnostics and therapeutics in oncology.

    Article  CAS  PubMed  Google Scholar 

  31. Graeber, T. G. et al. Hypoxia-mediated selection of cells with diminished apoptotic potential in solid tumours. Nature 379, 88–91 (1996).

    Article  CAS  PubMed  Google Scholar 

  32. Li, W. W. Tumour angiogenesis: molecular pathology, therapeutic targeting, and imaging. Acad. Radiol. 7, 800–811 (2000).A good overview of diagnostic opportunities in the field of angiogenesis.

    Article  CAS  PubMed  Google Scholar 

  33. Stohrer, M., Boucher, Y., Stangassinger, M. & Jain, R. K. Oncotic pressure in solid tumours is elevated. Cancer Res. 60, 4251–4255 (2000).

    CAS  PubMed  Google Scholar 

  34. Hansen-Algenstaedt, N. et al. Tumour oxygenation in hormone-dependent tumours during vascular endothelial growth factor receptor-2 blockade, hormone ablation, and chemotherapy. Cancer Res. 60, 4556–4560 (2000).This article provides an opportunity to explore the close correlation between angiogenesis and hypoxia and their modifications during therapeutic intervention.

    CAS  PubMed  Google Scholar 

  35. Abramovitch, R., Dafni, H., Smouha, E., Benjamin, L. E. & Neeman, M. In vivo prediction of vascular susceptibility to endothelial growth factor withdrawal: magnetic resonance imaging of C6 rat glioma in nude mice. Cancer Res. 59, 5012–5016 (1999).

    CAS  PubMed  Google Scholar 

  36. Pham, C. D. et al. Magnetic resonance imaging detects suppression of tumour vascular permeability after administration of antibody to vascular endothelial growth factor. Cancer Invest. 16, 225–230 (1998).

    Article  CAS  PubMed  Google Scholar 

  37. Taylor, N. J. et al. BOLD MRI of human tumour oxygenation during carbogen breathing. J. Magn. Reson. Imaging 14, 156–163 (2001).

    Article  CAS  PubMed  Google Scholar 

  38. Van Dijke, C. F. et al. Mammary carcinoma model: correlation of macromolecular contrast-enhanced MR imaging characterizations of tumour microvasculature and histologic capillary density. Radiology 198, 813–818 (1996).A description of the potential for contrast-enhanced MRI in the detailed description of the microvasculature.

    Article  CAS  PubMed  Google Scholar 

  39. Port, R. E. et al. Multicompartment analysis of gadolinium chelate kinetics: blood–tissue exchange in mammary tumours as monitored by dynamic MR imaging. J. Magn. Reson. Imaging 10, 233–241 (1999).

    Article  CAS  PubMed  Google Scholar 

  40. Tofts, P. S. et al. Quantitative analysis of dynamic Gd–DTPA enhancement in breast tumours using a permeability model. Magn. Reson. Med. 33, 564–568 (1995).

    Article  CAS  PubMed  Google Scholar 

  41. Brasch, R. C et al. In vivo monitoring of tumour angiogenesis with MR imaging. Acad. Radiol. 7, 812–823 (2000).

    Article  CAS  PubMed  Google Scholar 

  42. Jacquemier, J. et al. Angiogenesis as a prognostic marker in breast carcinoma with conventional adjuvant chemotherapy: a multiparametric and immunohistochemical analysis. J. Pathol. 184, 130–135 (1998).

    Article  CAS  PubMed  Google Scholar 

  43. Linderholm, B. et al. Vascular endothelial growth factor is of high prognostic value in node-negative breast carcinoma. J. Clin. Oncol. 16, 3121–3128 (1998).

    Article  CAS  PubMed  Google Scholar 

  44. Fuss, M. et al. Tumour angiogenesis of low-grade astrocytomas measured by dynamic susceptibility contrast-enhanced MRI (DSC–MRI) is predictive of local tumour control after radiation therapy. Int. J. Radiat. Oncol. Biol. Phys. 51, 478–482 (2001).

    Article  CAS  PubMed  Google Scholar 

  45. Padhani, A. R. et al. Effects of androgen deprivation on prostatic morphology and vascular permeability evaluated with MR imaging. Radiology 218, 365–374 (2001).

    Article  CAS  PubMed  Google Scholar 

  46. Brasch, R. et al. Assessing tumour angiogenesis using macromolecular MR imaging contrast media. J. Magn. Reson. Imaging 7, 68–74 (1997).

    Article  CAS  PubMed  Google Scholar 

  47. Ferrara, K. W. et al. Evaluation of tumour angiogenesis with US: imaging, Doppler, and contrast agents. Acad. Radiol. 7, 824–839 (2000).

    Article  CAS  PubMed  Google Scholar 

  48. Fleischer, A. C. Sonographic depiction of tumour vascularity and flow: from in vivo models to clinical applications. J. Ultrasound Med. 11, 377–385 (1992).

    Article  Google Scholar 

  49. Ismail, M., Peterson, R. O., Alexander, A. A., Newschaffer, C. & Gomella, L. G. Color Doppler imaging in predicting the biologic behavior of prostate cancer: correlation with disease-free survival. Urology 50, 906–912 (1997).

    Article  CAS  PubMed  Google Scholar 

  50. Lee, W. J. et al. Breast cancer vascularity: color Doppler sonography and histopathology study. Breast Cancer Res. Treat. 37, 291–298 (1996).

    Article  CAS  PubMed  Google Scholar 

  51. Donnelly, E. F., Geng, L., Wojcicki, W. E., Fleischer, A. C. & Hallahan, D. E. Quantified power Doppler US of tumour blood flow correlates with microscopic quantification of tumour blood vessels. Radiology 219, 166–170 (2001).

    Article  CAS  PubMed  Google Scholar 

  52. Rooks, V., Beecken, W.-D., Iordanescu, I. & Taylor, G. A. Sonographic evaluation of orthotopic bladder tumours in mice treated with TNP-470, an angiogenic inhibitor. Acad. Radiol. 8, 121–127 (2001).

    Article  CAS  PubMed  Google Scholar 

  53. Blankenberg, F. G. et al. Role of radionuclide imaging in trials of antiangiogenic therapy. Acad. Radiol. 7, 851–867 (2000).

    Article  CAS  PubMed  Google Scholar 

  54. Haubner, R. et al. Glycosylated RGD-containing peptides: tracer for tumour targeting and angiogenesis imaging with improved biokinetics. J. Nucl. Med. 42, 326–336 (2001).

    CAS  PubMed  Google Scholar 

  55. Miles, K. A. Tumour angiogenesis and its relation to contrast enhancement on computerised tomography: a review. Eur. J. Radiol. 30, 198–205 (1999).

    Article  CAS  PubMed  Google Scholar 

  56. Blomley, M. J. K. & Dawson, P. Bolus dynamics: theoretical and experimental aspects. Br. J. Radiol. 70, 351–359 (1997).

    Article  CAS  PubMed  Google Scholar 

  57. Cenic, A., Nabavi, D. G., Craen, R. A., Gelb, A. W. & Lee, T. Y. A CT method to measure hemodynamics in brain tumours: validation and application to cerebral blood flow maps. Am. J. Neuroradiol. 21, 462–470 (2000).

    CAS  PubMed  PubMed Central  Google Scholar 

  58. Shaheen, R. M. et al. Antiangiogenic therapy targeting the tyrosine kinase receptor for vascular endothelial growth factor receptor inhibits the growth of colon cancer liver metastasis and induces tumour and endothelial cell apoptosis. Cancer Res. 59, 5412–5416 (1999).

    CAS  PubMed  Google Scholar 

  59. Solorzano, C. C. et al. In vivo intracellular signaling as a marker of antiangiogenic activity. Cancer Res. 61, 7048–7051 (2001).

    CAS  PubMed  Google Scholar 

  60. Monestiroli, S. et al. Kinetics and viability of circulating endothelial cells as surrogate angiogenesis marker in an animal model of human lymphoma. Cancer Res. 61, 4341–4344 (2001).

    CAS  PubMed  Google Scholar 

  61. Poon, R. T.-P., Fan, S.-T. & Wang, J. Clinical implications of circulating angiogenic factors in cancer patients. J. Clin. Oncol. 19, 1207–1225 (2001).A comprehensive review of the role of circulating factors (mainly VEGF and bFGF) as prognostic and predictive markers in solid malignancies.

    Article  CAS  PubMed  Google Scholar 

  62. Millauer, B. et al. High affinity VEGF binding and developmental expression suggest Flk-1 as a major regulator of vasculogenesis and angiogenesis. Cell 72, 835–846 (1993).

    Article  CAS  PubMed  Google Scholar 

  63. Millauer B et al. Dominant-negative inhibition of Flk-1 suppresses the growth of many tumour types in vivo. Cancer Res. 56, 1615–1620 (1996).

    CAS  PubMed  Google Scholar 

  64. Rosen, L. S. et al. Phase I dose-escalating trial of SU5416, a novel angiogenesis inhibitor in patients with advanced malignancies. Proc. Am. Soc. Clin. Oncol. 18, 618 (1999).

  65. Gordon, et al. Phase I safety and pharmacokinetic study of recombinant human anti-vascular endothelial growth factor in patients with advanced cancer. J. Clin. Oncol. 19, 843–850 (2001).

    Article  CAS  PubMed  Google Scholar 

  66. Margolin, K. et al. Phase Ib trial of intravenous recombinant humanized monoclonal antibody to vascular endothelial growth factor in combination with chemotherapy in patients with advanced cancer: pharmacologic and long-term safety data. J. Clin. Oncol. 19, 851–856 (2001).

    Article  CAS  PubMed  Google Scholar 

  67. Rowinsky, E. K. et al. Phase I pharmacologic study of the specific metalloproteinase inhibitor BAY 12-9566 on a protacted oral daily dosing schedule in patients with solid malignancies. J. Clin. Oncol. 18, 178–186 (2000).

    Article  CAS  PubMed  Google Scholar 

  68. Gutheil, J. C. et al. Targeted antiangiogenic therapy for cancer using Vitaxin: a humanized monoclonal antibody to the integrin ανβ3 . Clin. Cancer Res. 6, 3056–3061 (2000).

    CAS  PubMed  Google Scholar 

  69. Boehle, A. S. et al. Human endostatin inhibits growth of human non-small cell lung cancer in a murine xenotransplant model. Int. J. Cancer 94, 420–428 (2001).

    Article  CAS  PubMed  Google Scholar 

  70. Fogler, W. E. et al. Recombinant human endostatin demonstrates consistent and predictable pharmacokinetics following intravenous bolus administration to cancer patients. Proc. Am. Soc. Clin. Oncol. 20, 274 (2001).

  71. Eder, J. P. et al. A Phase I pharmacokinetic and pharmacodynamic trial of recombinant human endostatin. Proc. Am. Soc. Clin. Oncol. 20, 275 (2001).

  72. Thomas, J. P. et al. A Phase I pharmacokinetic and pharmacodynamic study of recombinant human endostatin. Proc. Am. Soc. Clin. Oncol. 20, 276 (2001).

  73. Herbst, R. S. et al. Phase I clinical trial of recombinant human endostatin (rHE) in patients (Pts) with solid tumours: pharmacokinetic (PK), safety and efficacy analysis using surrogate endpoints of tissue and radiologic response. Proc. Am. Soc. Clin. Oncol. 20, 9 (2001).

  74. Davis, D. W. et al. Quantitative analysis of tumour vascular targeting: effects of endostatin in a Phase I clinical trial. J. Clin. Cancer Res. 7 (Suppl.), 3711 (2001).

    Google Scholar 

  75. Soker, S. et al. Circulating endothelial progenitor cells in cancer diagnosis and therapy. J. Clin. Cancer Res. 7 (Suppl.), 3665 (2001).

  76. Therasse, P. et al. New guidelines to evaluate the response to treatment in solid tumours. J. Natl Cancer Inst. 92, 205–216 (2000).A classic paper that every clinical investigator in cancer research should be familiar with.

    Article  CAS  PubMed  Google Scholar 

  77. Kopec, J. A., Abramowicz, M. & Esdail, J. M. Randomized discontinuation trials: utility and efficiency. J. Clin. Epidemiol. 46, 959–971 (1993).

    Article  CAS  PubMed  Google Scholar 

  78. Thall, P. F., Simon, R. M. & Estey, E. H. Bayesian sequential monitoring designs for single-arm trials with multiple outcomes. Stat. Med. 14, 357–379 (1995).An introduction to the complex topic of novel Bayesian monitoring methods applied to clinical-trial designs in oncology.

    Article  CAS  PubMed  Google Scholar 

  79. Ratain, M. J. et al. Clinical trial designs for cytotoxic agents J. Clin. Oncol. 19, 3154–3155 (2000).

    Article  Google Scholar 

  80. Rosner, G. L., Stadley, W. & Ratain, M. J. The randomized discontinuation design: application to cytotoxic antineoplastic agents. J. Clin. Oncol. (in the press).

  81. Kohn, E. C. et al. Structure–function analysis and growth inhibition by carboxyamido-triazole, CAI. Cancer Res. 54, 935–942 (1994).

    CAS  PubMed  Google Scholar 

  82. Bonomi, P. Matrix metalloproteinases and matrix metalloproteinase inhibitors in lung cancer. Semin. Oncol. 29, 78–86 (2002).

    Article  CAS  PubMed  Google Scholar 

  83. Coussens, L. M., Fingleton, B. & Matrisian, L. M. Matrix metalloproteinase inhibitors and cancer: trials and tribulations. Science 295, 2388–2392 (2002).

    Article  Google Scholar 

  84. Khokha, R. Suppression of the tumorigenic and metastatic abilities of murine B16-F10 melanoma cells in vivo by the overexpression of the tissue inhibitor of metalloproteinases-1. J. Natl Cancer Inst. 86, 299–304 (1994).

    Article  CAS  PubMed  Google Scholar 

  85. Gehan, E. A. & Freireich, E. J. Non-randomized controls in cancer clinical trials. N. Engl. J. Med. 290, 198–203 (1974).

    Article  CAS  PubMed  Google Scholar 

  86. Simon, R. Optimal two-stage designs for Phase II clinical trials. Control. Clin. Trials 10, 1–10 (1989).

    Article  CAS  PubMed  Google Scholar 

  87. Green, S. & Weiss, G. R. Southwest Oncology Group standard response criteria, endpoint definitions and toxicity criteria. Invest. New Drugs 10, 239–253 (1992).

    Article  CAS  PubMed  Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Massimo Cristofanilli.

Related links

Related links

DATABASES

Cancer.gov

AIDS-related Kaposi's sarcoma

brain tumour

breast cancer

cervical cancer

colon cancer

Kaposi's sarcoma

liver cancer

melanoma

multiple myeloma

non-Hodgkin's lymphoma

non-small-cell lung cancer

ovarian cancer

prostate cancer

renal-cell carcinoma

small-cell lung cancer

LocusLink

bFGF

collagen XVIII

COX-2

EGFR

FLK1

HER2

αv-integrin

β3-integrin

interferon-γ

IP-10

MMP2

MMP9

NF-κB

NHE3

p53

PDGF

phospholipase A2

phospholipase Cγ

PI3K

plasminogen

prostate-specific antigen

TNF-α

urokinase

VEGF

VEGFR2

Medscape DrugInfo

Gleevec

interferon-α

thalidomide

trastuzumab

FURTHER INFORMATION

European Organisation for Research and Treatment of Cancer

National Cancer Institute

Southwest Oncology Group

The Cancer and Leukemia Group B

The University of Texas M. D. Anderson Cancer Center

World Health Organization

Glossary

ANGIOPOIETINS

Angiopoietins are a novel family of proteins that specifically recognize and bind to the endothelial-cell-specific Tie2- receptor tyrosine kinase, and have been shown to be crucially involved in establishing the mature vascular network.

MICROVASCULATURE

A network of vessels that have diameters of less than 100 μm, which are beyond the resolution of conventional angiography.

MAGNETIC RESONANCE IMAGING

The use of radio waves in the presence of a magnetic field to extract information from certain atomic nuclei (most commonly hydrogen; for example, in water). Tissues can be differentiated by differences in their water densities. Tumours can be traced, as tumour tissue has a different water density from surrounding healthy tissue.

CONTRAST AGENTS

Contrast agents enhance the differences between normal and abnormal tissue in imaging studies.

ULTRASONOGRAPHY

The use of sound waves above the audible frequency to detect and characterize tumours. Echoes that are reflected off normal and abnormal tissues are captured by a computer to create two-dimensional images.

SINGLE-PHOTON-EMISSION COMPUTED TOMOGRAPHY

The detection and quantification of γ-emitting radionuclides, such as 99mTc, 111In, 123I or 125I.

POSITRON-EMISSION TOMOGRAPHY

An imaging technique that is used to detect decaying nuclides, such as 15O, 13N, 11C, 18F, 124I and 94mTc.

COMPUTED TOMOGRAPHY

A technique that exploits the differences in absorption of X-rays by different tissues to give high-contrast images of anatomical structures. Computed tomography has relatively poor soft-tissue contrast, so iodinated contrast agents, which perfuse different tissue types at different rates, are commonly used to delineate tumours.

GRADE III OR IV TOXICITIES

For each adverse event (AE) that is associated with a specific treatment, grades are assigned and defined using a scale from 0 to V. Grade III, severe and undesirable AE; grade IV, life threatening or disabling AE.

Rights and permissions

Reprints and permissions

About this article

Cite this article

Cristofanilli, M., Charnsangavej, C. & Hortobagyi, G. Angiogenesis modulation in cancer research: novel clinical approaches. Nat Rev Drug Discov 1, 415–426 (2002). https://doi.org/10.1038/nrd819

Download citation

  • Issue date:

  • DOI: https://doi.org/10.1038/nrd819

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing