7 Combinations of Cytotoxic Drugs,
Ionizing Radiation, and Angiogenesis Inhibitors
Carsten Nieder and Nicolaus H. Andratschke
C. Nieder, MD
N. H. Andratschke, MD
Department of Radiation Oncology, Klinikum rechts der Isar der Technischen Universität München, Ismaninger Strasse 22, 81675 München, Germany
7.1
Introduction
Many years after the fundamental hypotheses about delivery of oxygen and nutrients to malig- nant tumors via formation of new blood vessels (angiogenesis) were published, drugs developed to inhibit angiogenesis have now entered routine clinical practice, after landmark randomized trials have identified suitable combination regimens in metastatic colorectal cancer and advanced non- squamous non-small cell lung cancer (Table 7.1).
Previously, monotherapy, e.g., with matrix metallo- proteinase inhibitors, failed to improve the results.
The fundamental principles of anti-angiogenic approaches and their consequences for the devel-
opment and growth of solid tumors were published by Folkman (1971, 1986, 1995). Tumor angiogen- esis results from imbalance between pro-angiogenic factors, e.g., vascular endothelial growth factor (VEGF), fibroblast growth factor (FGF) and plate- let-derived growth factor (PDGF), and endogenous anti-angiogenic factors such as angiostatin and endostatin (Folkman 1995; O’Reilly et al. 1996, 1997; Endrich and Vaupel 1998; Ferrara and Alitalo 1999; Kerbel 2000; Carmeliet and Jain 2000; Yancopoulos et al. 2000). A large body of experimental data indicate that application of either inhibitors of pro-angiogenic factors or administra- tion of anti-angiogenic factors reduce the formation of new blood vessels. As a result, tumors grow at a slower rate or even decrease their size. However, in most cases no permanent tumor control can be achieved; therefore, the combination of anti-angio- genic strategies with cytotoxic agents such as che- motherapy, ionizing radiation, or both represents a promising approach to increase the cure rates of solid tumors ( Folkman 1971; Teicher et al. 1992;
Denekamp 1993; Folkman 1995; Siemann et al.
2000; Koukourakis 2001; Rosen 2002; Kal et al.
2004; Jain 2005; Xu et al. 2005). This chapter is focused on the VEGF pathways as an illustrative example of the therapeutic principles and efficacy.
Further data can be found in the disease-specific chapters such as 15.6.4 (pancreatic cancer) and 20.4.3 (gynecological cancers).
Angiogenesis is not restricted to tumors but can also be found in many other physiological and patho- logical conditions, e.g., normal growth, wound heal- ing, proliferative retinopathies, rheumatoid arthri- tis, and inflammation (for review see Folkman 1995; Carmeliet and Jain 2000); hence, inhibition of angiogenesis combined with other treatment may be associated with increased normal tissue reac- tions. In addition, several angiogenic growth factors were shown to reduce radiation-induced long-term toxicity, e.g., in the spinal cord and parotid glands (Andratschke et al. 2004, 2005; Thula et al. 2005);
therefore, the therapeutic ratio of anti-angiogenic
CONTENTS
7.1 Introduction 103
7.2 VEGFs in Tumor Angiogenesis 104 7.3 VEGF Inhibition Strategies 105 7.4 Combination of VEGF Inhibition with Radiotherapy 106
7.4.1 Tumor Models 106
7.4.2 Mechanisms Underlying the Enhanced Effect of Anti-VEGF Agents when Combined with Irradiation 108
7.4.3 Impact of Anti-VEGF Strategies on Radiobiological Hypoxia 108
7.4.4 Sequencing of Anti-VEGF Strategies Combined with Radiotherapy 109
7.4.5 Side Effects of Anti-VEGF Compounds Combined with Radiotherapy 109
7.5 Trimodal Treatment with Anti-Angiogenic Agents, Radiotherapy, and Chemotherapy 110
7.6 Conclusion 110 References 111
strategies in combination with radiotherapy needs to be thoroughly determined for safe translation of this approach into clinical practice.
7.2
VEGFs in Tumor Angiogenesis
VEGFs are potent mitogenes for endothelial cells and key mediators of tumor angiogenesis (for review see Dvorak et al. 1995; Kerbel et al. 1998; Ferrara 1999; Carmeliet 2000; Carmeliet and Collen 2000; Carmeliet and Jain 2000; Karkkainen and Petrova 2000; Kerbel 2000; Veikkola et al.
2000; Yancopoulos et al. 2000; Byrne et al. 2005).
The VEGFs represent a family of distinct proteins, e.g., VEGF (VEGF-A), VEGF-B, VEGF-C, VEGF-D, VEGF-E, and isoforms (e.g., VEGF121 and VEGF165).
Some family members are involved in lymphangio- genesis (VEGF-C and VEGF-D). Down-regulation of VEGF-C, e.g., by endostatin, might therefore inhibit lymph node metastasis (Fukumoto et al. 2005). The VEGF expression can be demonstrated in the vast majority of tumors and is usually elevated above normal tissue levels. Until tumors reach a volume of about 1 mm3, tumor cells are supplied by diffu- sion from the surrounding tissues (Folkman 1971;
Ausprunk and Folkman 1977; Folkman 1986). At larger volumes, impaired supply with oxygen and nutrients and accumulation of metabolites occur in the tumor. This is accompanied by important changes in the tumor microenvironment, e.g., hypoxia, hypoglycemia, and acidosis, which result in up-regulated production and release of HIF-1D, VEGF, and other angiogenic factors by the tumor cells (angiogenic switch, see Chap. 20.3.1 for further details). These factors represent potential targets
Table 7.1. Randomized clinical trials of chemotherapy plus/minus anti-angiogenic agents
Reference Compound Tumor Patient no. Results
Leighl et al.
(2005)
Paclitaxel/carboplatin plus BMS-275291 (MMPI) or placebo
NSCLC stage IIIB/IV 774 Outcome not improved, toxicity increased Bissett et al.
(2005)
Cisplatin/gemcitabine plus prinomastat (MMPI) or placebo
NSCLC stage IIIB/IV or recurrent
362 Outcome not improved, more treatment interruption Sparano et al.
(2004)
Marimastat (MMPI) or placebo after fi rst-line chemotherapy
Metastatic breast cancer
179 Outcome not improved, toxicity increased Bramhall et al.
(2002a)
Gemcitabine plus marimastat or placebo
Unresectable pancre- atic cancer
239 Outcome not improved, well tolerated
Bramhall et al.
(2002b)
Maintenance marimastat vs placebo after max. 1 5-FU-based chemotherapy regimen
Non-resectable gastric and gastro-esophageal cancer
369 2-year survival 3 vs 9%
in favor of marimastat (p=0.07), PFS sign. Longer Stadler et al.
(2004)
SU 5416 plus dexamethasone vs dexamethasone
Hormone-refractory prostate cancer
36 Outcome not improved
Szczylik et al.
(2005)
Sorafenib vs best supportive care Advanced renal cell carcinoma
769 PFS sign. improved (interim analysis)
Miller et al.
(2005)
Capecitabine plus/minus bevacizumab
Metastatic breast cancer
462 Sign. higher response rate (20 vs 9%), PFS, and OS not improved
Sandler et al.
(2005)
Paclitaxel/carboplatin plus/minus bevacizumab
Non-squamous NSCLC stage IIIB/IV
878 Sign. improvement in response rate, PFS and OS Kabbinavar et al.
(2005)
5-FU/LV plus bevacizumab or placebo
Metastatic colorectal cancer
209 PFS sign. better, OS better but not signifi cant Hurwitz et al.
(2004)
5-FU/LV/irinotecan plus placebo vs bevacizumab
Metastatic colorectal cancer
813 Sign. improvement in OS, PFS, and response (fi rst line) Giantonio et al.
(2005)
5-FU/LV/oxaliplatin (FOLFOX4) vs FOLFOX4 plus bevacizumab vs bevacizumab alone
Previously treated advanced colorectal cancer
829 Sign. improvement in PFS and OS
MMPI matrix metalloproteinase inhibitor, NSCLC non-small cell lung cancer, 5-FU 5-fl uorouracil, LV leucovorin, PFS progres- sion-free survival, OS overall survival
for anti-cancer treatment (Abdollahi et al. 2005;
Diaz-Gonzalez et al. 2005). In addition to envi- ronmental factors, other triggers, such as mechani- cal stress, immune/inflammatory responses, and genetic alterations (e.g., activation of oncogenes or deletion of tumor-suppressor genes that control pro- duction of angiogenesis regulators), may influence the expression of VEGF (Kerbel et al. 1998; Okada et al. 1998; Carmeliet and Jain 2000). Secreted VEGFs bind to specific receptors (VEGFR-1/Flt-1, VEGFR-2/KDR, VEGFR-3/Flt-4), which are almost exclusively expressed on the surface of endothelial cells (Jakeman et al. 1992; Veikkola et al. 2000;
Yancopoulos et al. 2000). Within the families of VEGFs and VEGFRs ligand affinity and recep- tor properties are variable. The VEGFRs consist of distinct structural elements, most importantly an extracellular immunoglobulin-like binding domain and an intracellular tyrosine kinase domain. Bind- ing of VEGF is followed by receptor dimerization, tyrosine kinase activation, autophosphorylation, and eventually activation of a complex cascade of intracellular pathways (Guo et al. 1995; Abedi and Zachary 1997; Karkkainen and Petrova 2000;
Veikkola et al. 2000; Stoletov et al. 2001). The activation of endothelial cells results in vasorelax- ation, increased vascular permeability, endothelial cell migration, proliferation, and survival. New blood vessels originating from surrounding, pre- existing normal vessels generate a tumor neovas- culature that allows further tumor growth, tumor progression, and development of metastasis; how- ever, important differences in vessel structure and organization exist, such as irregular leaky basement membranes (Di Tomaso et al. 2005).
7.3
VEGF Inhibition Strategies
Based on the molecular mechanisms underlying the biological effects of the VEGFs and their recep- tors, various rationally designed strategies to inhibit VEGF-dependent angiogenesis have been developed (Table 7.2). The best investigated approaches include antibodies against VEGF or VEGFR and VEGF receptor tyrosine kinase inhibitors (RTKI).
Table 7.2. Strategies to enhance the effi cacy of radiotherapy by inhibitors of angiogenesis
Target Mechanism Reference
VEGF ligand Neutralizing monoclonal antibodies Kim et al. (1993), Asano et al. (1995), Borgström et al. (1996), Presta et al. (1997)
Neutralizing chimeric proteins Aiello et al. (1995)
Soluble VEGFR-1 Lin et al. (1998), Goldman et al. (1998), Davidoff et al. (2001) Antisense oligonucleotides Bell et al. (1999), Im et al. (1999)
VEGF receptor Neutralizing monoclonal antibodies Prewett et al. (1999), Fenton et al. (2004), Li et al. (2005) Mutant VEGF receptor 2 (VEGFR-2) Machein et al. (1999)
Ribozyme targeted to mRNA Parry et al. (1999) Antisense oligonucleotides Rockwell et al. (1997)
Tyrosine kinase inhibitors Fong et al. (1999), Laird et al. (2000), Wood et al. (2000), Wedge et al. (2000), Zips et al. (2005)
Others Multitargeted kinase inhibitors, e.g., ZD6474 and SU11657
Williams et al. (2004), Damiano et al. (2005), Huber et al. (2005), Frederick et al. (2006)
Multitargeted growth factor-binding molecules
Sun et al. (2005)
Inhibitors of mTOR Shinohara et al. (2005; for further references see Chap. 9.4) Agents disrupting the tubulin cyto-
skeleton, e.g., ZD6126
Siemann and Rojiani (2005), Wachsberger et al. (2005)
Thalidomide Ansiaux et al. (2005)
HIV protease inhibitor ritonavir Maggiorella et al. (2005)
MN-029 Shi and Siemann (2005)
AMCA (lysine analog interfering with lysine binding sites of plasminogen)
Kal et al. (2004)
Metronomic chemotherapy Kerbel and Kamen (2004)
Neutralizing antibodies against VEGF or VEGFR2 can reduce the growth rate of tumors and metastases in experimental animals (Kim et al. 1993; Asano et al. 1995; Borgström et al. 1996; Presta et al. 1997;
Prewett et al. 1999; Gorski et al. 1999; Bruns et al.
2000; Lee et al. 2000; Kozin et al. 2001; Gupta et al.
2002; Winkler et al. 2004); however, considerable het- erogeneity in response to anti-VEGF or anti-VEGFR antibodies has been observed. In some tumors treated with anti-VEGF antibodies a nearly complete inhibi- tion of angiogenesis, a decline in vessel permeability, and a decrease in vascular density compared with control tumors has been demonstrated by intravital microscopy and histology (Borgström et al. 1996;
Yuan et al. 1996). Recent data suggest that VEGFR2 blockade increases tumor oxygenation, pericyte cov- erage of brain tumor blood vessels, and degradation of their pathologically thick basement membrane ( Winkler et al. 2004). To generate VEGF antibodies suitable for clinical trials, mouse monoclonal antibod- ies were humanized in order to combine high affin- ity to human VEGF with little or no immunogenicity (Presta et al. 1997; Ferrara and Alitalo 1999).
In a phase-I clinical trial including 25 patients with advanced solid tumors no dose-limiting toxicity up to the highest dose of recombinant human anti-VEGF monoclonal antibody (bevacizumab, 10 mg/kg body weight) was observed (Gordon et al. 2001); however, in this trial three episodes of tumor bleeding were noted. In two additional patients with known pulmo- nary metastasis minor hemoptysis occurred. It was not possible to discriminate whether these bleedings were caused by the treatment or by the underlying disease. In combination with chemotherapy rhumab- VEGF (3 mg/kg body weight) was safely administered without hemorrhagic complications ( Margolin et al.
2001). In a multicenter trial 99 patients with advanced non-small cell lung cancer were randomized to stan- dard chemotherapy or standard chemotherapy plus rhumabVEGF (DeVore et al. 2000). Response rate and time to progression were increased after combined treatment. In six patients, four of them with a cen- tral tumor location, severe tumor bleeding occurred, which was fatal in four patients.
VEGF-dependent angiogenesis can also be sup- pressed by RTKIs. Agents such as SU5416, ZD4190, and PTK787/ZK222584, competitively inhibit ATP- dependent phosphorylation of tyrosine residues of the VEGF receptor resulting in a decrease in prolifer- ation and survival of endothelial cells in vitro, angio- genesis in vivo, growth rate of tumor, and metastasis (Fong et al. 1999; Shaheen et al. 1999; Drevs et al.
2000; Laird et al. 2000; Mendel et al. 2000; Wedge
et al. 2000; Wood et al. 2000; Schuuring et al. 2005).
Although these compounds were designed to inhibit VEGFR2, some of them show also activity against other receptors involved in angiogenesis (Laird et al. 2000). In a clinical phase-I study on single-agent SU5416 treatment in patients with advanced malig- nant disease, the dose-limiting toxicity was vomit- ing, nausea, and severe headache (Cropp et al. 1999;
Rosen et al. 1999; Stopeck 2000). In a subsequent phase-I clinical trial SU5416 combined with cisplatin and gemcitabine in a total of 19 patients with solid tumors, an unexpectedly high rate of thrombem- bolic events was observed (Kuenen et al. 2002).
These discouraging results may be a consequence of the specific regimen of chemotherapy applied. This illustrates the importance of further, detailed inves- tigations of interactions and optimal scheduling of administration (Marx et al. 2002). Of special impor- tance in this context are pre-clinical investigations into normal tissue effects of combined approaches, exploiting suitable animal models.
7.4
Combination of VEGF Inhibition with Radiotherapy
7.4.1
Tumor Models
Whether anti-VEGF strategies in combination with radiotherapy can be used to improve tumor response was investigated in a number of experi- mental models (Table 7.3). In these studies different human and murine tumors, grown in experimental animals, were treated with various anti-VEGF com- pounds combined with single-dose or fractionated irradiation. Although VEGF inhibition alone had only a modest or no significant impact on tumor growth, the combination with irradiation consis- tently resulted in improved outcome. For example, in the study published by Gorski et al. (1999) anti-VEGF165 antibody alone did not reduce tumor growth rate in U87 human glioblastoma xenografts, whereas the combination of 40 Gy given in eight fractions with anti-VEGF165 antibody 3 h before each fraction increased tumor growth delay compared with irradiation alone. In another study performed by Kozin and colleagues (2001) two different human xenograft tumors were treated with an anti-VEGFR2 antibody combined with fractionated irradiation. In both tumor models the combined treatment resulted
in a statistically significant decrease of the dose nec- essary for permanent local tumor control compared with tumors that were only irradiated. This result demonstrates that anti-VEGFR antibody treatment increases the inactivation of clonogenic tumor cells and supports the hypothesis that anti-VEGF strate- gies may improve the outcome of curative radio- therapy; however, in the majority of the experi- ments listed in Table 7.3, tumor growth delay was
the experimental end point. Importantly, total dose, dose per fraction, overall treatment time, and the sequence of the modalities in combined treatment may impact on the outcome of combination treat- ment; hence, further pre-clinical studies in relevant animals models addressing these parameters need to be conducted to investigate whether the benefit of the combined therapy holds true for clinically relevant irradiation schedules.
Table 7.3. Pre-clinical in vivo studies testing anti-VEGF strategies in combination with radiotherapy. (Modifi ed from Zips and Baumann 2003)
Compound Mechanism of VEGF inhibition
Tumor Treatment schedule End point Reference
SU5416 RTKIa Murine SCC VIIb 5u2 Gy plus concomitant SU5416
Growth delay +c Ning et al.
(2002) SU6668 RTKI Murine SCC VII 5u2 Gy plus concomitant
SU6668
Growth delay + Ning et al.
(2002)
SU6668 RTKI Human mammary
carcinoma SCK
Two applications of SU6668 prior to irradiation with a single dose of 15 Gy
Growth delay + Griffi n et al.
(2002)
SU5416 RTKI Murine maligant
glioma GL261
Six fractions of 3 Gy with concomitant SU5416
Growth delay + Geng et al.
(2001) ZK 222584/
PTK 787
RTKI Human SW480 colon carcinoma
12 Gy in four fractions with concomitant ZK222584/
PTK787
Growth delay + Hess et al.
(2001)
A.4.6.1 Anti-VEGF antibody
Human glioblastoma U87, human colon adenocarcinoma LS174T
Single dose under normoxic or hypoxic conditions after antibody treatment
Growth delay + Lee et al.
(2000)
DC101 Anti-
VEGFR2 antibody
Human small cell lung cancer 54A, human glioblastoma U87
Five fractions in 5 days with six applications of DC101 every third day
Growth delay, tumor control rate
+ Kozin et al.
(2001)
DC101 Anti-
VEGFR2 antibody
Human SCC-1 or SCC-6 carcinoma
3 Gy twice weekly for 3.5 weeks with twice weekly DC101 for 3 weeks
Growth delay + Li et al.
(2005)
DC101 Anti-
VEGFR2 antibody
Mca-4 and Mca-35 mammary adenocar- cinoma
Five fractions of 6 Gy with DC101
Growth delay + Fenton et al.
(2004)
Anti-VEGF165 Anti-human VEGF165 antibody
Human SCC SQ20B, Seg-1 human adeno- carcinoma, human glioblastoma U87
Anti-VEGF165 prior to irradia- tion with four or eight frac- tions
Growth delay + Gorski et al.
(1999)
Anti-VEGF165 Anti-murine VEGF165 antibody
Murine Lewis lung carcinoma
40 Gy in two fractions with anti-VEGF165 prior to irradia- tion
Growth delay + Gorski et al.
(1999)
Anti-VEGF165 Anti-human VEGF165 antibody
Seg-1 human adeno- carcinoma, human glioblastoma U87
Anti-VEGF165 plus 20 Gy in four fractions (Seg-1) or 40 Gy in eight fractions (U87)
Growth delay + Gupta et al.
(2002)
aRTKI receptor tyrosine kinase inhibitor.
bSquamous cell carcinoma.
cBetter outcome with combined treatment compared to control tumor.
7.4.2
Mechanisms Underlying the Enhanced Effect of Anti-VEGF Agents when Combined with
Irradiation
The observation that anti-VEGF compounds alone only modestly impact tumor growth rate while in combination with irradiation anti-VEGF agents con- sistently improve the response of tumors, suggests a radiosensitizing effect. This might be restricted to endothelial cells, because VEGF receptors are almost exclusively expressed on this cell type (Jakeman et al. 1992; Ning et al. 2002). This notion is supported by the fact that neither unirradiated nor irradi- ated tumor cells were affected by anti-angiogenic compounds (Gorski et al. 1999; Hess et al. 2001;
Gupta et al. 2002; Dings et al. 2005). The VEGFs are potent mitogens and survival factors for endothe- lial cells (see section 7.2). Irradiation of endothelial cells in vitro results in decreased proliferation and decreased clonogenic survival (De Gowin et al. 1974;
Hei et al. 1987; Haimovitz-Friedman et al. 1991;
Fuks et al. 1994; Gorski et al. 1999; Hess et al. 2001;
Kermani et al. 2001; Gupta et al. 2002; Brieger et al. 2005). When VEGF is added to culture media, the effects of irradiation on proliferation and survival of endothelial cells were attenuated. On the other hand, VEGF inhibitors reduce these effects of VEGF on irradiated endothelial cells (Gorski et al. 1999;
Hess et al. 2001; Gupta et al. 2002). In studies using another angiogenic growth factor, bFGF, increased clonogenic survival of irradiated bovine aortic endo- thelial cells was observed (Haimovitz-Friedman et al. 1991; Fuks et al. 1994). In these studies the slope of the dose–survival curves was not significantly dif- ferent between endothelial cells incubated with or without bFGF; however, the shoulder region of the dose effect curve was almost completely eliminated when no bFGF was added to culture media. An inter- esting hypothesis of the mechanism of anti-VEGF combined with irradiation was proposed by Gorski and colleagues (1999) based on the fact that many cell types, including tumor cells, respond to irradiation with an increased release of growth factors such as VEGF or bFGF. These growth factors may in an auto- crine and/or paracrine manner increase endothelial cell survival after irradiation (Witte et al. 1989;
Haimovitz-Friedman et al. 1991; Gorski et al.
1999; Gupta et al. 2002). The authors hypothesized that the inhibition of VEGF can abrogate the effects of radiation-induced VEGF on irradiated endothe- lial cells in tumors. This may result in increased radiation-induced endothelial cell kill which even-
tually leads to impaired tumor vascularization. As a result, the tumor cell production rate decreases and/or the tumor cell loss increases which may result in improved tumor response after irradiation. Fur- ther experimental studies are required to elucidate the complex interactions of the endothelial cell and the tumor cell compartment after a combination of anti-angiogenic compounds with irradiation.
7.4.3
Impact of Anti-VEGF Strategies on Radiobiological Hypoxia
Theoretically, anti-angiogenic agents might increase tumor hypoxia. Anoxic cells are less sensitive to irra- diation compared with normoxic cells by a factor of about three (Gray et al. 1953; Wright and Howard- Flanders 1957); thus, inhibitors of angiogenesis, by decreasing vessel density or vessel function, might increase the proportion of poorly oxygenated clono- genic tumor cells and thereby decrease the efficacy of irradiation. A negative effect of anti-angiogenic agents on the results of irradiation was observed in an experiment by Murata and colleagues (1997).
The authors investigated the effect of TNP-470 on local tumor control after single-dose and fraction- ated irradiation of murine mammary carcinoma in C3H/He mice. The radiobiological hypoxic fraction of clonogenic tumor cells, as determined from the tumor control data after single-dose irradiation, was not affected significantly by TNP-470; however, for fractionated irradiation under normal blood flow conditions, tumors treated with TNP-470 were more resistant compared with control tumors. In contrast, no differences were observed for fractionated irra- diation under clamp hypoxia. The authors concluded that inhibition of angiogenesis by TNP-470 impairs reoxygenation during fractionated irradiation and thus increases radioresistance of tumors. In a study on human colon carcinoma growing in nude mice, another anti-angiogenic compound, suramine, was also found to increase radiobiological hypoxia (Leith et al. 1992). Yet, both TNP-470 (Zhang et al. 2000) and suramine do not specifically inhibit VEGF or VEGFR but decrease angiogenesis at least in part by other mechanisms. Whether anti-VEGF drugs similarly increase radiobiological hypoxia was addressed in two experimental studies using anti- VEGF and anti-VEGFR antibodies (Lee et al. 2000;
Kozin et al. 2001). Tumor growth delay after graded single radiation doses given under normal blood flow conditions was compared with tumor growth delay
after irradiations applied under clamp hypoxia. In both studies anti-VEGF and anti-VEGFR antibodies increased tumor growth delay compared with con- trol tumors irrespective of the tumor oyxgenation status at the time of irradiation. These results suggest that VEGF inhibition does not increase the radiobio- logical hypoxic cell fraction in tumors. The discrep- ancy of reduced vessel density on the one hand but lack of increase in hypoxia, on the other hand, may be attributed to an impact on the quality of vascu- lar organization by selective ablation of immature tumor blood vessels, a decrease in the number of oxygen consuming cells, and a decrease in vessel permeability (Benjamin et al. 1999; Lee et al. 2000).
Examination of FSAII tumors grown in mice demon- strated that anti-angiogenic treatment with thalido- mide caused reoxygenation within 2 days, accompa- nied by a reduction of interstitial fluid pressure and increase in perfusion (Ansiaux et al. 2005).
7.4.4
Sequencing of Anti-VEGF Strategies Combined with Radiotherapy
The VEGF inhibitors can be administrated before (neoadjuvant), during (simultaneous) and/or after (adjuvant) a course of fractionated radiotherapy. No conclusion on the optimal sequence can be drawn from the studies listed in Table 7.3. Current data indi- cate that VEGF inhibitors have only minor effect on tumor volume when given alone. Neoadjuvant appli- cation should have only little effect on the number of clonogenic tumor cells that need to be killed by irradiation; thus, a substantial improvement of the results of radiotherapy by neoadjuvant VEGF inhi- bition can only be expected from different mecha- nisms such as more functional vascularization and abrogation of resistance-inducing microenviron- mental parameters. Table 7.3 shows that simultane- ous application of VEGF inhibitors with fraction- ated irradiation improved the radiation response of several tumor models. These results might be caused by abrogation of the radioprotective effects of radia- tion-induced VEGF on endothelial cells. So far, the experimental results do not support that neoadju- vant or simultaneous application of VEGF inhibitors increases tumor hypoxia. To circumvent the possi- bility of increased hypoxia, anti-angiogenic agents may be applied after the end of fractionated irradia- tion. In a study by Murata and colleagues (1997) TNP-470 applied after a course of ten fractions in 2 weeks significantly increased tumor growth delay
compared with irradiation alone. In experiments by Zips et al. (2005), different combination schedules of a VEGFR tyrosine kinase inhibitor with fractionated irradiation of human squamous cell carcinomas in nude mice were compared. Short-term neoadjuvant and simultaneous administration showed no effect on tumor growth delay, whereas long-term adjuvant treatment resulted in prolonged tumor growth delay (but not increased cure rates). Arguments for adju- vant administration of VEGF inhibitors include the following: tumors may shrink after irradiation and smaller tumors are more likely to respond to anti- angiogenic compounds than larger tumors (Griffin et al. 2002). Irradiated vessels appear to be more sen- sitive to VEGF inhibition, which is supported by the observation that in vitro irradiated endothelial cells show an increased VEGFR2 expression (Kermani et al. 2001). An obvious disadvantage of the adju- vant sequence is that the radioprotective effect of VEGF on endothelial cells will not be counteracted.
Certainly, further studies to determine the optimal sequence of VEGF inhibition in combination with fractionated irradiation are needed.
7.4.5
Side Effects of Anti-VEGF Compounds Combined with Radiotherapy
In most of the studies listed in Table 7.3 the combi- nation with irradiation was well tolerated. Skin reac- tions after irradiation of the subcutaneously growing experimental tumors on the hind leg or flank of the animals were not increased; however, after whole- body irradiation in combination with anti-VEGFR2 antibody DC101 40% of 54A tumor-bearing mice developed ascites and half of them died (Kozin et al.
2001). Histology revealed focal segmental glomeru- lonecrosis associated with increased expression of VEGFR2 on the surface of glomerular endothelial cells in the kidneys (Kozin et al. 2002). This indi- cates that, although angiogenesis in adult organisms is almost exclusively confined to tumor tissue, inhi- bition of VEGF-dependent processes may result in increased normal tissue reactions after irradiation.
This might be explained by the fact that VEGF does not only stimulate angiogenesis but is also impor- tant for survival and maintenance of normal func- tion of endothelial cells. Such results underline the importance of further studies of normal tissue reac- tions, particularly those dominated by a long-term vascular component, after combined anti-VEGF strategies with radiotherapy.
7.5
Trimodal Treatment with Anti-Angiogenic Agents, Radiotherapy, and Chemotherapy
Kumar et al. (2005) reported that combination treatment with low doses of the phosphatidyl-ino- sitol 3-kinase (PI3K) inhibitor LY294002, cisplatin, and irradiation resulted in significantly higher endothelial cell death as compared with each agent used alone. This combination treatment was equally effective in inducing tumor cell death. Combination treatment also significantly inhibited endothelial cell tube formation in Matrigel as compared with each of the agents used alone. In an in vivo severe combined immunodeficient mouse model, combina- tion treatment with low doses of LY294002, cisplatin, and irradiation significantly inhibited the growth of human oral squamous carcinoma (OSCC-3) as well as prostate cancer (LnCap). The combina- tion therapy was also effective in inhibiting tumor angiogenesis where it showed a greater than 90%
decrease in neovascularization. In contrast, combi- nation treatment showed only a 29% inhibition of physiological angiogenesis.
Another group treated mice bearing Lewis lung carcinoma with combinations of intraperitoneal cisplatin, 5-fluorouracil, and the antiangiogenic agent genistein, together with 10 or 20 Gy of external beam radiotherapy (McDonnell et al. 2004). Ani- mals were sacrificed at day 6 when tumor volume, microvessel density, and serum VEGF were deter- mined. Mean tumor volume in the chemoradio- therapy group was 35% larger than in the chemo- radiotherapy plus genistein group (p=0.04). The addition of genistein produced a significant reduc- tion in tumor microvessel density as well as serum VEGF levels compared with those animals receiving chemoradiation alone.
Huber et al. (2005) found that in vitro and in vivo, the anti-endothelial and anti-tumor effects of the triple therapy combination consisting of SU11657 (a multitargeted small molecule inhibitor of VEGF and PDGF receptor tyrosine kinases), pemetrexed (a multitargeted folate antimetabolite), and ionizing radiation were superior to all single and dual com- binations. The superior effects in human umbilical vein endothelial cells and tumor cells (A431) were evident in cell proliferation, migration, tube for- mation, clonogenic survival, and apoptosis assays.
Triple therapy induced greater tumor growth delay than all other therapy regimens without increas- ing apparent toxicity. When testing different treat- ment schedules for the A431 tumor, these authors
found that the regimen with radiotherapy (7.5 Gy single dose), given after the institution of SU11657 treatment, was more effective than radiotherapy preceding SU11657 treatment. The SU11657 mark- edly reduced intratumoral interstitial fluid pressure after 1 day.
Among the multiple pathways altered by epi- dermal growth factor receptor (EGFR) inhibitors, angiogenesis appears to play an important role. Data on combined treatment with radiotherapy, chemo- therapy, and EGFR inhibitors are reviewed in Chap- ter 8.6.1.
7.6
Conclusion
Pre-clinical experiments in a variety of tumor models indicate that inhibition of angiogenic growth factors or growth factor receptors in combination with irra- diation is a promising concept for the improvement of the radiation response of tumors, although most data relate to tumor growth delay rather than cure.
The mechanisms underlying this approach are not fully understood to date. Both, radiosensitization of endothelial cells by VEGF inhibition as well as sensi- tization of endothelial cells against anti-angiogenic agents by irradiation appear to play an important role. In addition to approaches targeting VEGF and its pathways, a variety of other strategies are under investigation. Recently, evaluation of trimodal com- binations has started, because simultaneous radio- chemotherapy is now standard treatment for many types of cancer. Whether such strategies improve the therapeutic window remains to be determined.
So far, systematic data on long-term toxicity in many critical organs are lacking. Future research must include investigations of the efficacy of angiogenesis inhibition (and normalization of the microenviron- ment) in combination with irradiation and cytotoxic drugs with regard to dose-fractionation parameters, optimization of the dosing and sequence, identifica- tion of predictive factors for response, and studies on normal tissue toxicity in relevant experimental animal tumor and normal tissue models as well as early clinical trials. Examples of such trials in gas- trointestinal tumors are discussed in more detail in Chapter 15.6.4. In brief, preliminary experi- ence suggests that changes in tumor perfusion and interstitial intratumor pressure occur, and that acute toxicity of bevacizumab plus radiotherapy is acceptable. Further data have been published for
the combination of radiotherapy and temozolo- mide with thalidomide evaluated in a phase-II trial Chang et al. (2004). This regimen was administered to 67 patients with newly diagnosed glioblastoma. It was relatively well tolerated and showed a median survival of more than 16 months. Whether this represents an improvement over trials without tha- lidomide (median survival 1316 months) requires prospective confirmation.
References
Abdollahi A, Hlatky L, Huber PE (2005) Endostatin: the logic of antiangiogenic therapy. Drug Resist Update 8:59–74 Abedi H, Zachary I (1997) Vascular endothelial growth factor
stimulates tyrosine phosphorylation and recruitment to new focal adhesions of focal adhesion kinase and paxillin in endothelial cells. J Biol Chem 272:15442–15451
Aiello LP, Pierce EA, Foley ED et al (1995) Suppression of retinal neovascularization in vivo by inhibition of vas- cular endothelial growth factor (VEGF) using soluble VEGF-receptor chimeric proteins. Proc Natl Acad Sci USA 92:10457–10461
Andratschke NH, Nieder C, Price RE et al (2004) Modulation of rodent spinal cord radiation tolerance by administration of platelet-derived growth factor. Int J Radiat Oncol Biol Phys 60:1257–1263
Andratschke N, Nieder C, Price RE et al (2005) Potential role of growth factors in diminishing radiation therapy neural tissue injury. Semin Oncol 32 (Suppl 3):S67–S70
Ansiaux R, Baudelet C, Jordan BF et al (2005) Thalidomide radiosensitizes tumors through early changes in the tumor microenvironment. Clin Cancer Res 11:743–750
Asano M, Yukita A, Matsumoto T et al (1995) Inhibition of tumor growth and metastasis by an immunoneutraliz- ing monoclonal antibody to human vascular endothelial growth factor/vascular permeability factor121. Cancer Res 55:5296–5301
Ausprunk DH, Folkman J (1977) Migration and proliferation of endothelial cells in preformed and newly formed blood ves- sels during tumor angiogenesis. Microvasc Res 14:53–65 Bell C, Lynam E, Landfair DJ et al (1999) Oligonucleotide
NX1838 inhibits VEGF165-mediated cellular responses in vitro. In Vitro Cell Dev Biol Anim 35:533–542
Benjamin LE, Golijanin D, Itin A et al (1999) Selective ablation of immature blood vessels in established human tumors follows vascular endothelial growth factor withdrawal. J Clin Invest 103:159–165
Bissett D, O’Byrne KJ, Pawel J von et al (2005) Phase III study of matrix metalloproteinase inhibitor prinomastat in non- small-cell lung cancer. J Clin Oncol 23:842–849
Borgström P, Hillan KJ, Sriramarao P, Ferrara N (1996) Com- plete inhibition of angiogenesis and growth of microtu- mors by anti-vascular endothelial growth factor neutral- izing antibody: novel concepts of angiostatic therapy from intravital videomicroscopy. Cancer Res 56:4032–4039 Bramhall SR, Schulz J, Nemunaitis J et al (2002a) A double-
blind placebo-controlled, randomised study comparing
gemcitabine and marimastat with gemcitabine and placebo as first line therapy in patients with advanced pancreatic cancer. Br J Cancer 87:161–167
Bramhall SR, Hallissey MT, Whiting J et al (2002b) Marimastat as maintenance therapy for patients with advanced gastric cancer: a randomised trial. Br J Cancer 86:1864–1870 Brieger J, Schroeder P, Gosepath J, Mann WJ (2005) VEGF-sub-
type specific protection of SCC and HUVECs from radia- tion induced cell death. Int J Mol Med 15:145–151 Bruns CJ, Liu W, Davis DW et al (2000) Vascular endothelial
growth factor is an in vivo survival factor for tumor endo- thelium in a murine model of colorectal carcinoma liver metastases. Cancer 89:488–499
Byrne AM, Bouchier-Hayes DJ, Harmey JH (2005) Angiogenic and cell survival functions of vascular endothelial growth factor (VEGF). J Cell Mol Med 9:777–794
Carmeliet P (2000) Mechanisms of angiogenesis and arterio- genesis. Nat Med 6:389–395
Carmeliet P, Collen D (2000) Molecular basis of angiogen- esis. Role of VEGF and VE-cadherin. Ann NY Acad Sci 902:249–262
Carmeliet P, Jain RK (2000) Angiogenesis in cancer and other diseases. Nature 407:249–257
Chang SM, Lamborn KR, Malec M et al (2004) Phase II study of temozolomide and thalidomide with radiation therapy for newly diagnosed glioblastoma multiforme. Int J Radiat Oncol Biol Phys 60:353–357
Cropp G, Rosen L, Mulay M et al (1999) Pharmacokinetics and pharmacodynamics of SU5416 in a phase I, dose-escalating trial in patients with advanced malignancies. Proc Am Soc Clin Oncol 18:161a, abstact 619
Damiano V, Melisi D, Bianco C et al (2005) Cooperative anti- tumor effect of multitargeted kinase inhibitor ZD6474 and ionizing radiation in glioblastoma. Clin Cancer Res 11:5639–5644
Davidoff AM, Leary MA, Ng CY, Vanin EF (2001) Gene ther- apy-mediated expression by tumor cells of the angiogen- esis inhibitor flk-1 results in inhibition of neuroblastoma growth in vivo. J Pediatr Surg 36:30–36
De Gowin RL, Lewis LJ, Hoak JC et al (1974) Radiosensitiv- ity of human endothelial cells in culture. J Lab Clin Med 84:42–48
Denekamp J (1993) Review article: angiogenesis, neovascular proliferation and vascular pathophysiology as targets for cancer therapy. Br J Radiol 66:181–196
DeVore RF, Fehrenbacher L, Herbst RS et al (2000) A random- ized phase II trial comparing Rhumab VEGF (recombinant humanized monoclonal antibody to vascular endothelial growth factor) plus carboplatin/paclitaxel (CP) to CP alone in patients with stage IIIB/IV NSCLC. Proc Am Soc Clin Oncol 19:454a. Abstract 1896
Diaz-Gonzalez JA, Russell J, Rouzaut A et al (2005) Targeting hypoxia and angiogenesis through HIF-1alpha Inhibition.
Cancer Biol Ther 4:1055–1062
Dings RP, Williams BW, Song CW et al (2005) Anginex syn- ergizes with radiation therapy to inhibit tumor growth by radiosensitizing endothelial cells. Int J Cancer 115:312–
319
Drevs J, Hofmann I, Hugenschmidt H et al (2000) Effects of PTK787/ZK 222584, a specific inhibitor of vascular endo- thelial growth factor receptor tyrosine kinases, on primary tumor, metastasis, vessel density, and blood flow in a murine renal cell carcinoma model. Cancer Res 60:4819–4824
Dvorak HF, Brown LF, Detmar M, Dvorak AM (1995) Vascu- lar permeability factor/vascular endothelial growth factor, microvascular hyperpermeability, and angiogenesis. Am J Pathol 146:1029–1039
Endrich B, Vaupel P (1998) The role of the microcirculation in the treatment of malignant tumors: facts and fiction.
In: Molls M, Vaupel P (eds) Blood perfusion and microen- vironment of human tumors. Springer, Berlin Heidelberg New York, pp 19–39
Fenton BM, Paoni SF, Ding I (2004) Pathophysiological effects of vascular endothelial growth factor receptor-2-blocking antibody plus fractionated radiotherapy on murine mam- mary tumors. Cancer Res 64:5712–5719
Ferrara N (1999) Molecular and biological properties of vascu- lar endothelial growth factor. J Mol Med 77:527–543 Ferrara N, Alitalo K (1999) Clinical applications of angiogenic
growth factors and their inhibitors. Nat Med 5:1359–1364 Folkman J (1971) Tumor angiogenesis: therapeutic implica-
tions. N Engl J Med 285:1182–1186
Folkman J (1986) How is blood vessel growth regulated in normal and neoplastic tissue? G.H.A. Clowes Memorial Award lecture. Cancer Res 46:467–473
Folkman J (1995) Angiogenesis in cancer, vascular, rheuma- toid and other disease. Nat Med 1:27–31
Fong TA, Shawver LK, Sun L et al (1999) SU5416 is a potent and selective inhibitor of the vascular endothelial growth factor receptor (Flk-1/KDR) that inhibits tyrosine kinase catalysis, tumor vascularization, and growth of multiple tumor types. Cancer Res 59:99–106
Frederick B, Gustafson D, Bianco C et al (2006) ZD6474, an inhibitor of VEGFR and EGFR tyrosine kinase activity in combination with radiotherapy. Int J Radiat Oncol Biol Phys 64:33–37
Fuks Z, Persaud RS, Alfieri A et al (1994) Basic fibroblast growth factor protects endothelial cells against radia- tion-induced programmed cell death in vitro and in vivo.
Cancer Res 54:2582-2590
Fukumoto S, Morifuji M, Katakura Y et al (2005) Endostatin inhibits lymph node metastasis by a down-regulation of the vascular endothelial growth factor C expression in tumor cells. Clin Exp Metastasis 22:31–38
Geng L, Donnelly E, McMahon G et al (2001) Inhibition of vascular endothelial growth factor receptor signaling leads to reversal of tumor resistance to radiotherapy. Cancer Res 61:2413–2419
Giantonio BJ, Catalano PJ, Meropol NJ et al (2005) High- dose bevacizumab improves survival when combined with FOLFOX4 in previously treated advanced colorec- tal cancer: results from the Eastern Cooperative Oncol- ogy Group (ECOG) study E3200 (Abstract). J Clin Oncol 23:1s
Goldman CK, Kendall RL, Cabrera G et al (1998) Paracrine expression of a native soluble vascular endothelial growth factor receptor inhibits tumor growth, metastasis, and mortality rate. Proc Natl Acad Sci USA 95:8795–8800 Gordon MS, Margolin K, Talpaz M et al (2001) 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
Gorski DH, Beckett MA, Jaskowiak NT et al (1999) Blockage of the vascular endothelial growth factor stress response increases the antitumor effects of ionizing radiation.
Cancer Res 59:3374–3378
Gray LH , Conger AD, Ebert M et al (1953) The concentration of oxygen dissolved in tissues at the time of irradiation as a factor in radiotherapy. Br J Radiol 26:638–648
Griffin RJ, Williams BW, Wild R et al (2002) Simultane- ous inhibition of the receptor kinase activity of vascular endothelial, fibroblast, and platelet-derived growth factors suppresses tumor growth and enhances tumor radiation response. Cancer Res 62:1702–1706
Guo D, Jia Q, Song HY et al (1995) Vascular endothelial cell growth factor promotes tyrosine phosphorylation of medi- ators of signal transduction that contain SH2 domains.
Association with endothelial cell proliferation. J Biol Chem 270:6729–6733
Gupta VK, Jaskowiak NT, Beckett MA et al (2002) Vascular endothelial growth factor enhances endothelial cell sur- vival and tumor radioresistance. Cancer J 8:47–54 Haimovitz-Friedman A, Vlodavsky I, Chaudhuri A et al. (1991)
Autocrine effects of fibroblast growth factor in repair of radiation damage in endothelial cells. Cancer Res 51:2552–
2558
Hei TK, Marchese MJ, Hall EJ (1987) Radiosensitivity and sub- lethal damage repair in human umbilical cord vein endo- thelial cells. Int J Radiat Oncol Biol Phys 13:879–884 Hess C, Vuong V, Hegyi I et al (2001) Effect of VEGF recep-
tor inhibitor PTK787/ZK222584 [correction of ZK222548]
combined with ionizing radiation on endothelial cells and tumour growth. Br J Cancer 85:2010–2016
Huber PE, Bischof M, Jenne J et al (2005) Trimodal cancer treatment: beneficial effects of combined antiangiogenesis, radiation, and chemotherapy. Cancer Res 65:3643–3655 Hurwitz H, Fehrenbacher L, Novotny W et al (2004) Bevaci-
zumab plus irinotecan, fluorouracil, and leucovorin for metastatic colorectal cancer. N Engl J Med 350:2335–2342 Im SA, Gomez-Manzano C, Fueyo J et al (1999) Antiangiogen-
esis treatment for gliomas: transfer of antisense-vascular endothelial growth factor inhibits tumor growth in vivo.
Cancer Res 59:895–900
Jain RK (2005) Normalization of tumor vasculature: an emerging concept in antiangiogenic therapy. Science 307:58–62
Jakeman LB, Winer J, Bennett GL et al (1992) Binding sites for vascular endothelial growth factor are localized on endo- thelial cells in adult rat tissues. J Clin Invest 89:244–253 Kabbinavar FF, Schulz J, McCleod M et al (2005) Addition of
bevacizumab to bolus fluorouracil and leucovorin in first- line metastatic colorectal cancer: results of a randomized phase II trial. J Clin Oncol 23:3697–3705
Kal HB, Struikmans H, Gebbink MF, Voest EE (2004) Response of rat prostate and lung tumors to ionizing radiation com- bined with the angiogenesis inhibitor AMCA. Strahlenther Onkol 180:798–804
Karkkainen MJ, Petrova TV (2000) Vascular endothelial growth factor receptors in the regulation of angiogenesis and lymphangiogenesis. Oncogene 19:5598–5605
Kerbel RS (2000) Tumor angiogenesis: past, present and the near future. Carcinogenesis 21:505–515
Kerbel RS, Kamen BA (2004) The anti-angiogenic basis of met- ronomic chemotherapy. Nat Rev Cancer 4:423–436 Kerbel RS, Viloria-Petit A, Okada F, Rak J (1998) Establishing
a link between oncogenes and tumor angiogenesis. J Mol Med 4:286–295
Kermani P, Leclerc G, Martel R, Fareh J (2001) Effect of ion- izing radiation on thymidine uptake, differentiation, and
VEGFR2 receptor expression in endothelial cells: the role of VEGF(165). Int J Radiat Oncol Biol Phys 50:213–220 Kim KJ, Li B, Winer J, Armanini M et al (1993) Inhibition of
vascular endothelial growth factor-induced angiogenesis suppresses tumour growth in vivo. Nature 362:841–844 Koukourakis MI (2001) Tumour angiogenesis and response to
radiotherapy. Anticancer Res 21:4285–4300
Kozin SV, Boucher Y, Hicklin DJ et al (2001) Vascular endo- thelial growth factor receptor-2-blocking antibody potenti- ates radiation-induced long-term control of human tumor xenografts. Cancer Res 6:39–44
Kozin SV, Boucher Y, Tomaso E di et al (2002) Glomerulopathy in mice treated with whole-body irradiation and VEGFR2- blocking antibody. 49th Annual Meeting of the Radiation Research Society, Reno, Nevada, 2002, P08–P65
Kuenen BC, Rosen L, Smit EF et al (2002) Dose-finding and pharmacokinetic study of cisplatin, gemcitabine, and SU5416 in patients with solid tumors. J Clin Oncol 20:1657–
1667
Kumar P, Benedict R, Urzua F et al (2005) Combination treat- ment significantly enhances the efficacy of antitumor therapy by preferentially targeting angiogenesis. Lab Invest 85:756–767
Laird AD, Vajkoczy P, Shawver LK et al (2000) SU6668 is a potent antiangiogenic and antitumor agent that induces regres- sion of established tumors. Cancer Res 60:4152–4160 Lee CG, Heijn M, Tomaso E di et al (2000) Anti-vascular endo-
thelial growth factor treatment augments tumor radiation response under normoxic or hypoxic conditions. Cancer Res 60:5565–5570
Leighl NB, Paz-Ares L, Douillard JY et al (2005) Randomized phase III study of matrix metalloproteinase inhibitor BMS- 275291 in combination with paclitaxel and carboplatin in advanced non-small-cell lung cancer: National Cancer Institute of Canada Clinical Trials Group Study BR.18. J Clin Oncol 23:2831–2839
Leith JT, Papa G, Quaranto L, Michelson S (1992) Modifica- tion of the volumetric growth responses and steady-state hypoxic fractions of xenografted DLD-2 human colon carcinomas by administration of basic fibroblast growth factor or suramin. Br J Cancer 66:345–348
Li J, Huang S, Armstrong EA et al (2005) Angiogenesis and radiation response modulation after vascular endothelial growth factor receptor-2 (VEGFR2) blockade. Int J Radiat Oncol Biol Phys 62:1477–1485
Lin P, Sankar S, Shan S et al (1998) Inhibition of tumor growth by targeting tumor endothelium using a soluble vascular endothelial growth factor receptor. Cell Growth Differ 9:49–58
Machein MR, Risau W, Plate KH (1999) Antiangiogenic gene therapy in a rat glioma model using a dominant-negative vascular endothelial growth factor receptor 2. Hum Gene Ther 10:1117–1128
Maggiorella L, Wen B, Frascogna V et al (2005) Combined radiation sensitizing and anti-angiogenic effects of ioniz- ing radiation and the protease inhibitor ritonavir in a head and neck carcinoma model. Anticancer Res 25:4357–4362 Margolin K, Gordon MS, Holmgren E et al. (2001) Phase Ib
trial of intravenous recombinant humanized monoclonal antibody to vascular endothelial growth factor in combina- tion with chemotherapy in patients with advanced cancer:
pharmacologic and long-term safety data. J Clin Oncol 19:851–856
Marx GM, Steer CB, Harper P et al (2002) Unexpected serious toxicity with chemotherapy and antiangiogenic combina- tions: time to take stock! J Clin Oncol 20:1446–1448 McDonnell CO, Holden G, Sheridan ME et al (2004) Improve-
ment in efficacy of chemoradiotherapy by addition of an antiangiogenic agent in a murine tumor model. J Surg Res 116:19–23
Mendel DB, Schreck RE, West DC et al (2000) The angiogen- esis inhibitor SU5416 has long-lasting effects on vascular endothelial growth factor receptor phosphorylation and function. Clin Cancer Res 6:4848–4858
Miller KD, Chap LI, Holmes FA et al (2005) Randomized phase III trial of capecitabine compared with bevacizumab plus capecitabine in patients with previously treated metastatic breast cancer. J Clin Oncol 23:792–799
Murata R, Nishimura Y, Hiraoka M (1997) An antiangiogenic agent (TNP-470) inhibited reoxygenation during fraction- ated radiotherapy of murine mammary carcinoma. Int J Radiat Oncol Biol Phys 37:1107–1113
Ning S, Laird D, Cherrington JM, Knox SJ (2002) The antian- giogenic agents SU5416 and SU6668 increase the antitumor effects of fractionated irradiation. Radiat Res 157:45–51 Okada F, Rak JW, Croix BS et al (1998) Impact of oncogenes
in tumor angiogenesis: mutant K-ras up-regulation of vascular endothelial growth factor/vascular permeability factor is necessary, but not sufficient for tumorigenicity of human colorectal carcinoma cells. Proc Natl Acad Sci USA 95:3609–3614
O’Reilly MS, Holmgren L, Chen C, Folkman J (1996) Angio- statin induces and sustains dormancy of human primary tumors in mice. Nat Med 2:689–692
O’Reilly MS, Boehm T, Shing Y et al (1997) Endostatin: an endogenous inhibitor of angiogenesis and tumor growth.
Cell 88:277–285
Parry TJ, Cushman C, Gallegos AM et al (1999) Bioactivity of anti-angiogenic ribozymes targeting Flt-1 and KDR mRNA.
Nucleic Acids Res 27:2569–257
Presta LG, Chen H, O’Connor SJ et al (1997) Humanization of an anti-vascular endothelial growth factor monoclonal antibody for the therapy of solid tumors and other disor- ders. Cancer Res 57:4593–4599
Prewett M, Huber J, Li Y et al (1999) Antivascular endothelial growth factor receptor (fetal liver kinase 1) monoclonal antibody inhibits tumor angiogenesis and growth of sev- eral mouse and human tumors. Cancer Res 59:5209–5218 Rockwell P, O’Connor WJ, King K et al (1997) Cell-surface
perturbations of the epidermal growth factor and vascular endothelial growth factor receptors by phosphorothioate oligodeoxynucleotides. Proc Natl Acad Sci USA 94:6523–
6528
Rosen LS (2002) Clinical experience with angiogenesis signal- ing inhibitors: focus on vascular endothelial growth factor (VEGF) blockers. Cancer Control 9 (2 Suppl):36–44 Rosen L, Mulay M, Mayers A et al (1999) Phase I dose-esca-
lating trial of SU5416, a novel angiogenesis inhibitor in patients with advanced malignancies. Proc Am Soc Clin Oncol 18:161a, abstract 618
Sandler AB, Gray R, Brahmer J et al (2005) Randomized phase II/III trial of paclitaxel (P) plus carboplatin (C) with or without bevacizumab in patients with advanced non- squamous non-small cell lung cancer (NSCLC): an East- ern Cooperative Oncology Group (ECOG) trial – E4599 (Abstract). J Clin Oncol 23:2s
Schuuring J, Bussink J, Bernsen H et al (2005) Irradiation combined with SU5416: microvascular changes and growth delay in a human xenograft glioblastoma tumor line. Int J Radiat Oncol Biol Phys 61:529–534
Shaheen RM, Davis DW, Liu W et al (1999) Antiangiogenic therapy targeting the tyrosine kinase receptor for vascular endothelial growth factor receptor inhibits the growth of colon cancer liver metastasis and induces tumor and endo- thelial cell apoptosis. Cancer Res 59:5412–5416
Shi W, Siemann DW (2005) Preclinical studies of the novel vas- cular disrupting agent MN-029. Anticancer Res 25:3899–
3904
Shinohara ET, Cao C, Niermann K et al (2005) Enhanced radi- ation damage of tumor vasculature by mTOR inhibitors.
Oncogene 24:5414–5422
Siemann DW, Rojiani AM (2005) The vascular disrupting agent ZD6126 shows increased antitumor efficacy and enhanced radiation response in large, advanced tumors. Int J Radiat Oncol Biol Phys 62:846–853
Siemann DW, Warrington KH, Horsman MR (2000) Target- ing tumor blood vessels: an adjuvant strategy for radiation therapy. Radiother Oncol 57:5–12
Sparano JA, Bernardo P, Stephenson P et al (2004) Ran- domized phase III trial of marimastat versus placebo in patients with metastatic breast cancer who have respond- ing or stable disease after first-line chemotherapy: East- ern Cooperative Oncology Group trial E2196. J Clin Oncol 22:4683–4690
Stadler WM, Cao D, Vogelzang NJ et al (2004) A randomized phase II trial of the antiangiogenic agent SU5416 in hor- mone-refractory prostate cancer. Clin Cancer Res 10:3365–
3370
Stoletov KV, Ratcliffe KE, Spring SC, Terman BI (2001) NCK and PAK participate in the signaling pathway by which vas- cular endothelial growth factor stimulates the assembly of focal adhesions. J Biol Chem 276:22748–22755
Stopeck A (2000) Results of a phase I dose-escalating study of the antiangiogenic agent, SU5416, in patients with advanced malignancies. Proc Am Soc Clin Oncol 19:206a, abstract 802
Sun J, Wang DA, Jain RK et al (2005) Inhibiting angiogenesis and tumorigenesis by a synthetic molecule that blocks binding of both VEGF and PDGF to their receptors. Onco- gene 24:4701–4709
Szczylik C, Eisen T, Stadler WM et al (2005) Randomized phase III trial of the Raf kinase and VEGFR inhibitor sorafenib (BAY 43-9006) in patients with advanced renal cell carci- noma (Abstract). J Clin Oncol 23:1093s
Teicher BA, Sotomayor EA, Huang ZD (1992) Antiangiogenic agents potentiate cytotoxic cancer therapies against pri- mary and metastatic disease. Cancer Res 52:6702–6704 Thula TT, Schultz G, Tran-Son-Tay R, Batich C (2005) Effects of
EGF and bFGF on irradiated parotid glands. Ann Biomed Eng 33:685–695
Tomaso E di, Capen D, Haskell A et al (2005) Mosaic tumor ves- sels: cellular basis and ultrastructure of focal regions lack- ing endothelial cell markers. Cancer Res 65:5740–5749
Veikkola T, Karkkainen M, Claesson-Welsh L, Alitalo K (2000) Regulation of angiogenesis via vascular endothelial growth factor receptors. Cancer Res 60:203–212
Wachsberger PR, Burd R, Marero N et al (2005) Effect of the tumor vascular-damaging agent, ZD6126, on the radiore- sponse of U87 glioblastoma. Clin Cancer Res 11:835–842 Wedge SR, Ogilvie DJ, Dukes M et al (2000) ZD4190: an orally
active inhibitor of vascular endothelial growth factor sig- naling with broad-spectrum antitumor efficacy. Cancer Res 60:970–975
Williams KJ, Telfer BA, Brave S et al (2004) ZD6474, a potent inhibitor of vascular endothelial growth factor signaling, combined with radiotherapy: schedule-dependent enhance- ment of antitumor activity. Clin Cancer Res 10:8587–8593 Winkler F, Kozin SV, Tong RT et al (2004) 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 Witte L, Fuks Z, Haimovitz-Friedman A et al (1989) Effects of
irradiation on the release of growth factors from cultured bovine, porcine, and human endothelial cells. Cancer Res 49:5066–5072
Wood JM, Bold G, Buchdunger E et al.(2000) PTK787/ZK 222584, a novel and potent inhibitor of vascular endo- thelial growth factor receptor tyrosine kinases, impairs vascular endothelial growth factor-induced responses and tumor growth after oral administration. Cancer Res 60:2178–2189
Wright EA, Howard-Flanders P (1957) The influence of oxygen on the radiosensitivity of mammalian tissues. Acta Radiol 48:26–32
Xu L, Tong R, Cochran DM, Jain RK (2005) Blocking plate- let-derived growth factor-D/platelet-derived growth factor receptor beta signaling inhibits human renal cell carci- noma progression in an orthotopic mouse model. Cancer Res 65:5711–5719
Yancopoulos GD, Davis S, Gale NW et al (2000) Vascular-spe- cific growth factors and blood vessel formation. Nature 407:242–248
Yuan F, Chen Y, Dellian M et al (1996) Time-dependent vascular regression and permeability changes in established human tumor xenografts induced by an anti-vascular endothelial growth factor/vascular permeability factor antibody. Proc Natl Acad Sci USA 93:14765–14770
Zhang Y, Griffith EC, Sage J et al (2000) Cell cycle inhibition by the anti-angiogenic agent TNP-470 is mediated by p53 and p21WAF1/CIP1. Proc Natl Acad Sci USA 97:6427–6432 Zips D, Baumann M (2003) Anti-VEGF strategies in combi-
nation with radiotherapy. In: Nieder C, Milas L, Ang KK (eds) Modification of radiation response. Springer, Berlin Heidelberg New York, pp 179–188
Zips D, Hessel F, Krause M et al (2005) Impact of adjuvant inhibition of vascular endothelial growth factor receptor tyrosine kinases on tumor growth delay and local tumor control after fractionated irradiation in human squamous cell carcinomas in nude mice. Int J Radiat Oncol Biol Phys 61:908–914