Monitoring Changes in the Microenvironment During Targeted Therapies

Publication
Article
OncologyONCOLOGY Vol 21 No 11
Volume 21
Issue 11

This review covers progress to date in the identification of molecular targets on blood vessels in cancers, as well as agents that act on those targets, with emphasis on those currently in clinical trials. Current vascular-targeting therapies comprise two general types—antiangiogenic therapy and antivascular therapy. Advances in antiangiogenic therapies, particularly inhibitors of vascular endothelial growth factors and their receptors, have clarified the capacity of these inhibitors to change tumor-associated vessel structure to a more normal state, thereby improving the ability of chemotherapeutics to access the tumors. The responses of other antiangiogenesis target molecules in humans are more complicated; for example, αvβ3 integrins are known to stimulate as well as inhibit angiogenesis, and cleavage of various extracellular proteins/proteoglycans by matrix metalloproteinases produces potent regulators of the angiogenic process. Antivascular therapies disrupt established blood vessels in solid tumors and often involve the use of ligand-based or small-molecule agents. Ligand-based agents, irrespective of the antiangiogenic capacity of the ligand, target antivascular effectors to molecules expressed specifically on blood vessels, such as aminopeptidase N, fibronectin extra-domain B, and prostate-specific membrane antigen. Small-molecule antivascular agents, which are not targeted to molecules on blood vessels, rely on physical differences between the vasculatures in tumors and those in normal tissues.

Tumor angiogenesis has been a subject of wide interest over the past decade, following from the groundbreaking work of Folkman and colleagues.[1] Uncontrolled angiogenesis is present in almost every tumor type and leads to immature, malformed vessels lined by endothelial cells with aberrant morphology, loosely attached pericytes, and an abnormal basal membrane. These vessels are characterized by their chaotic organization and increased permeability causing elevated interstitial fluid pressure.[2] Vascular malformations do not provide an efficient supply of oxygen, nutrients, or therapeutic drugs to tumor tissue. This leads to significant treatment resistance.[3,4]

In this issue of ONCOLOGY, Sato et al provide a comprehensive overview of different strategies for antiangiogenic and antivascular targeted therapies, focusing on those currently in clinical trials. This commentary will focus on the need for methods to monitor changes in the microenvironment and angiogenic responses to vascular-targeted therapies, alone and in combination with cytotoxic therapies.

Considerations When Using Antiangiogenic Agents

Antiangiogenesis was first considered as a treatment option based on the hope that it would destroy tumor vasculature to the point that tumors regressed as a result of severe oxygen starvation. While this approach seems theoretically attractive, that outcome has not proven to be the case for antiangiogenic therapies.

In fact, correcting the overexpression of proangiogenic factors (such as vascular endothelial growth factor [VEGF]) by antiangiogenic compounds leads to a better balance of pro- vs antiangiogenic factors. This can lead to maturation and/or pruning of immature vessels, causing "normalization" of tumor vasculature.[5-10] Of utmost importance in "normalization" is whether oxygen and drug delivery are enhanced, and if so, the kinetics of the responses.

The concept of "vascular normalization" has been evaluated at the preclinical level in numerous studies, briefly summarized below. A murine mammary carcinoma (McaIV) and a human colon carcinoma cell line (LS174T) showed less tortuous vessels 3 days after blocking VEGF receptor (VEGFR)2 with the murine VEGF antibody DC101. Remaining vessels were more uniformly covered by pericytes and basement membrane.[10] These normalized vessels were less leaky, as reflected by a reduction of intratumoral fluid pressure, and there was improvement in oxygenation, as assessed by hypoxia marker drug uptake. Vessel normalization was followed by vessel regression at day 5, however, demonstrating the transient nature of this response.[10] In a neuroblastoma (NB-1691) xenograft model, VEGF inhibiton by bevacizumab (Avastin) led to better intratumoral drug delivery and efficacy. Both bevacizumab and DC101 improved tumor tissue oxygenation[9,11] in human glioma xenografts, resulting in increased efficacy of radiation treatment.[9]

Small-molecule inhibitors of VEGF receptor activity have also shown promise.[12] After 4 days of treatment of a non–small-cell lung cancer (NSCLC) xenograft tumor line with imatinib (Gleevec), a platelet-derived growth factor receptor (PDGFR) inhibitor, investigators observed a significant reduction of VEGF expression and vascular normalization (defined by tight pericyte-endothelial cell coverage). In addition, they noted improved oxygenation after 1 to 2 weeks of therapy and an enhanced antitumor effect when imatinib was combined with docetaxel.[13,14]

Not all reports have been positive with respect to whether normalization occurs after initiation of VEGF inhibition, however. Fenton et al reported variable results in different tumor lines and suggested that direct sensitization of tumor endothelial cells to radiation therapy following withdrawal of VEGF signaling might be more important in explaining radiosensitization than any change in oxygenation.[15] The mechanism of treatment failure of individual tumors is not fully understood. One possible explanation is the ability of certain tumor cells to dedifferentiate and build vessel-like-structures-the so-called vasculogenic mimicry. This process of cell plasticity occurs mainly in highly aggressive tumors and was first discovered in highly metastatic melanoma. These vessels seem not to respond to antiangiogenic treatment.[16]

Clinically, Willett et al performed serial studies in patients with locally advanced rectal cancer to monitor the effects of bevacizumab, prior to initiation of radiotherapy. They observed reductions in perfusion (assessed by contrast computed tomography [CT]), interstitial fluid pressure, microvessel density[17] and circulating levels of endothelial cell precursors.[18] Another extensive study in patients with glioblastoma multiforme was reported recently by the same group. Using dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI) methods, they observed significant reductions in intratumoral edema and microvessel diameter response to a small-molecule VEGFR inhibitor.[19] Although these measures strongly suggest that oxygenation of the tumors improved after antiangiogenic therapy, there have not been any clinical reports examining hypoxia as an endpoint, which is crucial for making treatment decisions that involve oxygen-dependent therapies, such as radiation and chemotherapy.

 

Influence of Cytotoxic Therapies on VEGF and Angiogenesis

It is important to recognize that VEGF levels in tumors change in response to various forms of cancer therapies and that these changes influence vascular responsiveness and endothelial cell survival after therapy. An important regulator of VEGF activity in tumors is the hypoxia-inducible transcription factor (HIF-1), which binds to the promoter region of VEGF to stimulate transcription.[20] HIF-1 is active predominantly under hypoxic conditions, because its alpha subunit is targeted by the proteosome under normoxic conditions for hydroxylation reactions in its oxygen-dependent degradation domain, modifying it for recognition by the VHL complex and subsequent ubiquitinylation and proteosomal degradation.

Cytotoxic treatment of tumors often leads to transient improvement in oxygenation, in a process originally coined "reoxygenation" by Rubin and Casarett.[21] One might think that reoxygenation would lead to reduced HIF-1 activity, but in fact, the opposite effect occurs. During the process of reoxygenation, HIF-1 levels actually increase, largely in response to production of free radicals that prevent the oxygen-dependent degradation of HIF-1.[22,23] The increase in HIF-1 activity increases VEGF levels, thereby protecting them from damage by radiation or chemotherapy. In effect, this indirectly protects the tumor, by preserving its vasculature. This provides a strong rationale for inhibiting VEGF during radiotherapy or chemotherapeutic treatment.

Teicher was the first to show that inhibition of angiogenesis led to improved chemotherapy and radiotherapy response in several different tumor lines.[24] Gorski et al were the first to demonstrate that VEGF levels increased after radiotherapy and further demonstrated that inhibition of VEGF increased radioresponse.[25] Our group was the first to show that elevations in HIF-1 after radiotherapy were responsible for increases in VEGF[22] and that blockade of HIF-1 upregulation led to profound vascular sensitization to radiotherapy.

 

Vascular Targeting

In contrast to the "normalization effects"of antiangiogenic approaches, vascular-targeting therapies more closely approximate the original goal of antiangiogenesis. They rapidly shut down tumor perfusion, leading to tumor cell starvation and death. With this form of therapy, the ability to image treatment responses is very clear. The pure vascular-targeting agents, such as combretastatin A4, cause profound reductions in tumor perfusion within minutes of treatment, yet when used alone, have virtually no effect on tumor growth. This is because the highly proliferative tumor margin is able to repopulate the tumor quite readily.[26] The need for imaging to monitor vascular responses and demonstrate bioactivity with such agents is virtually mandatory in the absence of significant tumor volume reduction.

 

Biomarkers of Activity

The discussion above illustrates the importance of biomarkers of relevant physiologic parameters for phase I/II trials of antiangiogenic and vascular-targeting agents. Markers of bioactivity of such agents can be broken down into three categories: (1) imaging-based, (2) blood-based, and (3) tissue-based markers. It is preferable to include biomarkers that reflect perfusion, permeability, and oxygenation or drug transport.

• Imaging-Based Biomarkers-The relative merits of different MR- or CT-based imaging biomarkers are outside the realm of this commentary, but we will emphasize the need to develop methods that can distinguish changes in permeability from changes in vascular surface area. Typical small-molecule contrast agents cannot do this. An alternative would be to use macromolecular contrast agents or blood pool agents that show limited transvascular transport.[27,28]

In addition, one should not ignore the importance of monitoring hypoxia. The oxygenation state of a tumor is of pivotal importance in influencing treatment outcome from either radiation or chemotherapy. Since both antiangiogenic and vascular-targeting therapies can influence oxygenation, this information is pivotal in establishing how to combine such therapies with radiation or chemotherapy. Several positron-emission tomography (PET) hypoxia-imaging reagents are under development that could be used in this context, but to our knowledge none have been. Examples include 18F misonidazole,[29] 18F EF5,[30,31] and CuATSM.[32]

Some investigators have advocated the use of so-called blood oxygen level detection (BOLD) imaging to monitor changes in vascular oxygenation in tumors. The method relies on changes in MRI properties of hemoglobin in different oxygenation states. The problem with this method for this particular application is that BOLD depends on the differentiation of images at baseline and after the patient breathes a hyperoxic gas. Such gases are vasoactive, leading to changes in perfusion and microvessel hematocrit, both of which independently influence the BOLD signal.[33]

• Blood-Based Biomarkers-Recently, two plasma-based markers of hypoxia have been shown to be correlated with tumor hypoxia. Plasminogen activator inhibitor (PAI)-1 is an extracelluar protein involved in stabilizing matrix. It is regulated by HIF-1 and can be detected in plasma. Osteopontin is another secreted protein involved in stimulating cell motility and metastasis. It is regulated by hypoxia via a Ras-mediated enhancer. Both of these proteins have been strongly correlated with the presence of hypoxia in tumors, based on independent direct measurements of hypoxia, such as by Eppendorf polarographic electrodes.[34-37]

• Invasive and Tissue-Based Biomarkers-The Eppendorf electrode is a polarographic needle device that can be used to measure tumor hypoxia.[38] Although this is a highly validated method for performing such measurements, the device is cumbersome, particularly in situations where multiple measurements are required, as would be needed in a monitoring trial.

Similarly, several endogenous protein markers, alone or in combination, have been correlated with invasive measurements of hypoxia and have been shown to be prognostically important.[34,39] However, they suffer from the same fate as the Eppendorf electrode, in that multiple biopsies would be required for monitoring. Because of the heterogeneity of tumor hypoxia, this would likely require multiple biopsies at each time point, which would not be feasible for most trials.

 

Summary

Both antiangiogenic and vascular-targeted therapies hold great promise. The approval of some antiangiogenic agents in combination with other therapies bode well for the future clinical use of these agents. Similarly, the increased interest in vascular-targeting approaches shows potential, particularly when combined with radiotherapy and/or chemotherapy. In both of these approaches, however, it is imperative that suitable biomarkers be employed to monitor treatment effects and ensure optimal use. To achieve the highest efficacy of these treatment modalities, it may be mandatory to determine the response of antiangiogenic and antivascular compounds to ascertain an appropriate treatment schedule on an individualized basis (see Figure 1).

 

-Melanie Wergin, DVM, PHD
-Christopher G. Willett, MD
-Mark W. Dewhirst, DVM, PHD

Disclosures:

The authors have no significant financial interest or other relationship with the manufacturers of any products or providers of any service mentioned in this article.

References:

1. Folkman J: Angiogenesis: An organizing principle for drug discovery? Nat Rev Drug Discov 6:273-286, 2007.

2. Ferrara N: Vascular endothelial growth factor as a target for anticancer therapy. Oncologist 9(suppl 1):2-10, 2004.

3. Brizel DM, Rosner GL, Harrelson J, et al: Pretreatment oxygenation profiles of human soft tissue sarcomas. Int J Radiat Oncol Biol Phys 30:635-642, 1994.

4. Brizel DM, Rosner GL, Prosnitz LR, et al: Patterns and variability of tumor oxygenation in human soft tissue sarcomas, cervical carcinomas, and lymph node metastases. Int J Radiat Oncol Biol Phys 32:1121-1125, 1995.

5. Fukumura D, Jain RK: Tumor microvasculature and microenvironment: Targets for anti-angiogenesis and normalization. Microvasc Res May 18, 2007 (epub ahead of print).

6. Jain RK: Normalization of tumor vasculature: An emerging concept in antiangiogenic therapy. Science 307:58-62, 2005.

7. Jain RK: Molecular regulation of vessel maturation. Nat Med 9:685-693, 2003.

8. Jain RK, Tong RT, Munn LL: Effect of vascular normalization by antiangiogenic therapy on interstitial hypertension, peritumor edema, and lymphatic metastasis: Insights from a mathematical model. Cancer Res 67:2729-2735, 2007.

9. Winkler F, Kozin SV, Tong RT, et al: Kinetics of vascular normalization by VEGFR2 blockade governs brain tumor response to radiation: Role of oxygenation, angiopoietin-1, and matrix metalloproteinases. Cancer Cell 6:553-563, 2004.

10. Tong RT, Boucher Y, Kozin SV, et al: Vascular normalization by vascular endothelial growth factor receptor 2 blockade induces a pressure gradient across the vasculature and improves drug penetration in tumors. Cancer Res 64:3731-3736, 2004.

11. Lee CG, Heijn M, di Tomaso E, et al: Anti-vascular endothelial growth factor treatment augments tumor radiation response under normoxic or hypoxic conditions. Cancer Res 60:5565-5570, 2000.

12. Brazelle WD, Shi W, Siemann DW: VEGF-associated tyrosine kinase inhibition increases the tumor response to single and fractionated dose radiotherapy. Int J Radiat Oncol Biol Phys 65:836-841, 2006.

13. Vlahovic G, Ponce AM, Rabbani Z, et al: Treatment with imatinib improves drug delivery and efficacy in NSCLC xenografts. Br J Cancer 97:735-740, 2007.

14. Vlahovic G, Rabbani ZN, Herndon JE 2nd, et al: Treatment with imatinib in nsclc is associated with decrease of phosphorylated PDGFR-beta and VEGF expression, decrease in interstitial fluid pressure and improvement of oxygenation. Br J Cancer 95:1013-1019, 2006.

15. Fenton BM, Paoni SF, Grimwood B, et al: Varied response of spontaneous tumors to antiangiogenic agents. Adv Exp Med Biol 566:59-65, 2005.

16. Folberg R, Maniotis AJ: Vasculogenic mimicry. APMIS 112:508-525, 2004.

17. 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 23:8136-8139, 2005.

18. Duda DG, Cohen KS, di Tomaso E, et al: Differential CD146 expression on circulating versus tissue endothelial cells in rectal cancer patients: Implications for circulating endothelial and progenitor cells as biomarkers for antiangiogenic therapy. J Clin Oncol 24:1449-1453, 2006.

19. Batchelor TT, Sorensen AG, di Tomaso E, et al: AZD2171, a pan-VEGF receptor tyrosine kinase inhibitor, normalizes tumor vasculature and alleviates edema in glioblastoma patients. Cancer Cell 11:83-95, 2007.

20. Semenza GL: Targeting HIF-1 for cancer therapy. Nat Rev Cancer 3:721-732, 2003.

21. Rubin P, Casarett G: Microcirculation of tumors. II. The supervascularized state of irradiated regressing tumors. Clin Radiol 17:346-355, 1966.

22. Moeller BJ, Cao Y, Li CY, et al: Radiation activates HIF-1 to regulate vascular radiosensitivity in tumors: Role of reoxygenation, free radicals, and stress granules. Cancer Cell 5:429-441, 2004.

23. Li F, Sonveaux P, Rabbani ZN, et al: Regulation of HIF-1alpha stability through S-nitrosylation. Mol Cell 26:63-74, 2007.

24. Teicher BA, Sotomayor EA, Huang ZD: Antiangiogenic agents potentiate cytotoxic cancer therapies against primary and metastatic disease. Cancer Res 52:6702-6704, 1992.

25. Gorski DH, Beckett MA, Jaskowiak NT, et al: Blockage of the vascular endothelial growth factor stress response increases the antitumor effects of ionizing radiation. Cancer Res 59:3374-3378, 1999.

26. Tozer GM, Kanthou C, Baguley BC: Disrupting tumour blood vessels. Nat Rev 5:423-435, 2005.

27. Persigehl T, Bieker R, Matuszewski L, et al: Antiangiogenic tumor treatment: Early noninvasive monitoring with USPIO-enhanced MR imaging in mice. Radiology 244:449-456, 2007.

28. Preda A, van Vliet M, Krestin GP, et al: Magnetic resonance macromolecular agents for monitoring tumor microvessels and angiogenesis inhibition. Invest Radiol 41:325-331, 2006.

29. Rajendran JG, Schwartz DL, O'Sullivan J, et al: Tumor hypoxia imaging with [F-18] fluoromisonidazole positron emission tomography in head and neck cancer. Clin Cancer Res 12:5435-5441, 2006.

30. Dolbier WR Jr, Li AR, Koch CJ, et al: [18F]-EF5, a marker for PET detection of hypoxia: Synthesis of precursor and a new fluorination procedure. Appl Radiat Isot 54:73-80, 2001.

31. Evans SM, Kachur AV, Shiue CY, et al: Noninvasive detection of tumor hypoxia using the 2-nitroimidazole [18F]EF1. J Nucl Med 41:327-336, 2000.

32. Lewis JS, Welch MJ: PET imaging of hypoxia. Q J Nucl Med 45:183-188, 2001.

33. Neeman M, Dafni H, Bukhari O, et al: In vivo bold contrast MRI mapping of subcutaneous vascular function and maturation: Validation by intravital microscopy. Magn Reson Med 45:887-898, 2001.

34. Cecic I, Chan DA, Sutphin PD, et al: Oxygen sensitivity of reporter genes: Implications for preclinical imaging of tumor hypoxia. Mol Imaging 6:219-228, 2007.

35. Le QT, Chen E, Salim A, et al: An evaluation of tumor oxygenation and gene expression in patients with early stage non-small cell lung cancers. Clin Cancer Res 12:1507-1514, 2006.

36. Le QT, Kong C, Lavori PW, et al: Expression and prognostic significance of a panel of tissue hypoxia markers in head-and-neck squamous cell carcinomas. Int J Radiat Oncol Biol Phys 69:167-175, 2007.

37. Le QT, Sutphin PD, Raychaudhuri S, et al: Identification of osteopontin as a prognostic plasma marker for head and neck squamous cell carcinomas. Clin Cancer Res 9:59-67, 2003.

38. Tatum JL, Kelloff GJ, Gillies RJ, et al: Hypoxia: Importance in tumor biology, non-invasive measurement by imaging, and value of its measurement in the management of cancer therapy. Int J Radiat Biol 82:699-757, 2006.

39. Kim SJ, Rabbani ZN, Dewhirst MW, et al: Expression of HIF-1alpha, CA IX, VEGF, and MMP-9 in surgically resected non-small cell lung cancer. Lung Cancer 49:325-335, 2005.

Related Videos
A panel of 4 experts on lung cancer
A panel of 4 experts on lung cancer
A panel of 3 experts on multiple myeloma
A panel of 3 experts on multiple myeloma
A panel of 3 experts on multiple myeloma
Aparna Parikh, MD, with the Oncology Brothers presenting slides
Aparna Parikh, MD, with the Oncology Brothers presenting slides