Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2009 May 4.
Published in final edited form as: Cancer Metastasis Rev. 2008 Sep;27(3):415–434. doi: 10.1007/s10555-008-9143-x

Clinical biomarkers of angiogenesis inhibition

Aaron P Brown 1, Deborah E Citrin 2, Kevin A Camphausen 3
PMCID: PMC2676436  NIHMSID: NIHMS101783  PMID: 18414993

Abstract

Introduction

An expanding understanding of the importance of angiogenesis in oncology and the development of numerous angiogenesis inhibitors are driving the search for biomarkers of angiogenesis. We review currently available candidate biomarkers and surrogate markers of anti-angiogenic agent effect.

Discussion

A number of invasive, minimally invasive, and non-invasive tools are described with their potential benefits and limitations. Diverse markers can evaluate tumor tissue or biological fluids, or specialized imaging modalities.

Conclusions

The inclusion of these markers into clinical trials may provide insight into appropriate dosing for desired biological effects, appropriate timing of additional therapy, prediction of individual response to an agent, insight into the interaction of chemotherapy and radiation following exposure to these agents, and perhaps most importantly, a better understanding of the complex nature of angiogenesis in human tumors. While many markers have potential for clinical use, it is not yet clear which marker or combination of markers will prove most useful.

Keywords: Cancer, Angiogenesis, Biomarker, Imaging

1 Background

Angiogenesis, defined as the formation of new blood vessels, is a necessary process for tissue survival in physiologic and pathologic states. The study of angiogenesis has rapidly expanded since Judah Folkman first suggested in 1971 that angiogenic dysregulation could be required for tumor growth and metastasis [1]. Angiogenesis has since become an accepted target for anti-cancer therapy [25]. In 2003, bevacizumab became the first angiogenesis inhibitor to be approved by the FDA for use in the U.S. Currently there are several anti-angiogenic agents in clinical use or in testing for cancer therapy as well as many others that exhibit anti-angiogenic properties as part of their mechanism of action (Table 1). There are currently over 1,000 interventional clinical trials investigating over 40 anti-angiogenic agents in cancer treatment (Table 2, www. clinicaltrials.gov).

Table 1.

FDA-approved drugs with presumed anti-angiogenic mechanisms

Category Drug name Target/mechanism Cancer indications Current clinical trialsa
References
Phase (no.) Tumor or histology
Monoclonal antibodies Bevacizumab (Avastin) VEGF mCRC, NSCLC I (58);
 II (336);
 III (51);
 IV (5)
Numerous [271274]
Cetuximab (Erbitux) EGFR mCRC, H & N I (3); II (18);
 III (4) IV (1)
H & N, RCC, lung,
 pancreas, CRC, glioma
[275, 276]
Panitumumab
 (Vectibix)
EGFR mCRC II (2); III (1) CRC, lung [277, 278]
Trastuzumab
 (Herceptin)
EGFR, HER-2 Breast Cancer I (3); II (8);
 III (2)
Breast, endometrial,
 gastric, lung, ovarian
[279]
Small-molecule tyrosine kinase
 inhibitors
Erlotinib (Tarceva) EGFR NSCLC, Pancreatic Cancer I (16); II (44);
 III (4)
Numerous [280, 281]
Gefitinib (Iressa) EGFR NSCLC II (3) Skin, RCC [282]
Imatinib (Gleevec) bcr/abl, c-kit,
 PDGFR
CML, GIST I (5); II (15);
 III (9);
 IV (1)
GIST, leukemia,
 melanoma, lung, prostate,
 liposarcoma
[283, 284]
Lapatinib (Tykerb) EGFR, HER-2 Breast Cancer I (1); II (1) Breast, solid tumors [285]
Sorafenib (Nexavar) VEGFR-2, -3,
 PDGFR-ß, Raf-1,
RCC I (3); II (9);
 III (3)
GIST, melanoma,
 fallopian ovarian, RCC
[271]
Sunitinib (Sutent) VEGFR-2, PDGFR-ß GIST, RCC I (27); II (85);
 III (17)
Numerous [286, 287]
Other agents with anti-
 angiogenic properties
Bortezomib (Velcade) Proteosome Inhibitor MM, Mantle Cell Lymphoma I (7); II (16);
 III (6)
MM, lung, pancreas, RCC,
 lymphoma
[288, 289]
Interferon
 Alfa-2b (Roferon A,
 Intron A)
Inhibits angiogenic
 factor expression
Hairy cell leukemia,
 CML, NHL, Melanoma,
 Kaposi’s Sarcoma,
I (36);
II (126);
III (52);
 IV (3)
Numerous [290, 291]
Temsirolimus (Torisel) mTOR inhibitor RCC I (22); II (36);
 III (4)
Numerous [287, 292]
Thalidomide
 (Thalomid)
Unknown MM I (29);
 II (149);
 III (39)
Numerous [293]
Lenalidomide
 (Revlamid)
Unknown MM I (9); II (19);
 III (5)
Numerous [294]

mCRC Metastatic colorectal cancer, NSCLC non-small cell lung cancer, H & N head and neck cancer, CML chronic myelogenous leukemia, GIST gastrointestinal stromal tumor, RCC renal cell carcinoma, MM multiple myeloma, NHL non-Hodgkin’s lymphoma

Table 2.

Presumed anti-angiogenic agents currently in clinical trialsa

Category Drug name Target/mechanism Current clinical trialsa
Phase
(no.)
Cancers in trials
Target angiogenic factor Volociximab
 (M200)
Anti-α5β1 integrin monoclonal
 antibody
II Melanoma
Aflibercept (VEGF
 TrapR1/R2)
VEGF decoy receptor II, III Ovary, lung
PI-88 Mimics Heparan Sulfate, inhibits
 heparanase, binds FGF and VEGF
II Prostate
PTC299 VEGF 5′-UTR mRNA I Breast
Ad-hIFN-β
 (BG00001)
IFN-β gene, adenovirus vector II CNS, CRC, mesothelioma
Target angiogenic signal:
 tyrosine kinase inhibitors
Cediranib
 (AZD2171)
VEGF I, II,
 III
CRC, solid tumors, lymphoma
Nilotinib
 (AMN107)
KIT, PDGFR, and Bcr-Abl III GIST
Vatalanib
 (PTK787/ZK
 222584)
VEGF I, II,
 III
CRC, MM, metastatic tumors
AMG 706 VEGF, PDGF, c-kit, Ret II, III Breast, NSCLC
Pazopanib
 (GW786034)
VEGFR, c-kit, PDGF-R II, III RCC
Axitinib (AG-
 013736)
VEGFR-1, -2, PDGF-βR I, II Lung, thyroid, CRC, melanoma,
 kidney, pancreas
Semaxanib
 (SU5416)
Flk-1/KDR I, II Fallopian tube, breast, H & N,
 sarcoma, melanoma, kidney
AG-6013736 VEGFR, PDGFR II Thyroid
Brivanib (BMS-
 582664)
VEGFR, FGFR II HCC
ABT-869 VEGFR, PDGFR II RCC
Dasatinib (BMS-
 354825)
bcr/abl I Solid tumors
Vandetanib (ZD
 6474)
VEGFR, EGFR I-III Numerous
RAF265 Raf, VEGFR-2 I Metastatic melanoma
Target endothelial cells TNP-470 Fumagillin analogue, binds MetAP2
 causing EC arrest in G1 phase
I, II Pancreas, Kaposi’s sarcoma
Squalamine lactate Intracellular blockage of numerous
 pathways
II Prostate, ovarian
Recombinant tPA Induces Angiostatin II Solid tumors
Ad-rhEndo (E10A) Endostatin gene transfer I Advanced Solid tumor
ABT-510 Thrombospondin Analogue: inhibits
 VEGF, bFGF, HGF, IL-8
I Solid tumors
Col-3 MMP inhibitor I Metastatic cancer
2-methoxyestradiol
 (Panzem)
Estradiol Metabolite I, II RCC, carcinoid, solid tumors
Combretastatin A4
 Phosphate
Tubulin I Solid tumors
Bavituximab Aminophospholipids monoclonal
 antibody
I Solid tumors
Other agents with anti-
 angiogenic properties
AMG 386 Angiopoietins II Breast
Tetrathiomolybdate Copper Chelator II Breast, prostate, CRC, HCC,
 esophageal
Celecoxib COX-2 inhibitor I-III MM, head and neck, CRC,
 leukemia, lymphoma

mCRC Metastatic colorectal cancer, NSCLC non-small cell lung cancer, H & N head and neck cancer, CML chronic myelogenous leukemia, GIST gastrointestinal stromal tumor, RCC renal cell carcinoma, MM multiple myeloma, NHL Non-Hodgkin’s lymphoma

Angiogenesis is a complex process with numerous potential therapeutic targets. In cancer, angiogenesis is initiated when a tumor cell produces a pro-angiogenic signal, or angiogenic factor, activating resting endothelial and stromal cells. The activated endothelial cells then acquire the ability to remodel adjacent extracellular matrix, proliferate, migrate, then differentiate and stabilize as new blood vessels [6, 7]. Known angiogenic factors include vascular endothelial growth factor (VEGF), platelet derived growth factor (PDGF), hepatocyte growth factor (HGF), basic fibroblast growth factor (bFGF), matrix metalloproteinases (MMPs), transforming growth factor (TGF) -β, interleukin (IL) -8 and angiopoietins [4]. The most well-characterized and most commonly targeted pathway is VEGF and its tyrosine kinase receptors (VEGFR-1 and -2) [8, 9].

The majority of the agents in use or in development target angiogenic factor pathways or the endothelial cell (Tables 1 and 2; reviewed in [6, 10]). The most common target in anti-angiogenic therapy is disruption of the angiogenic signal. This can be done in many ways. First, the angiogenic factor itself can be targeted by either direct inhibition, such as with bevacizumab, a monoclonal antibody to VEGF-A, or by creating a sink for the factor, exemplified by VEGF-Trap (aflibercept), a soluble form of VEGFR. Inhibiting angiogenic factors is possible by decreasing production, (i.e. interferon (IFN) -α, COX-2 inhibitors) or activity (i.e. suramin). Toxins bound to angiogenic factors are another potential anti-angiogenic method currently in preclinical studies [11, 12]. Targeting the receptor also disrupts the angiogenic signal, shown by cetuximab, a monoclonal antibody to the epidermal growth factor receptor (EGFR). Several drugs act on signaling pathways, specifically by targeting the tyrosine kinase receptors of angiogenic factors (i.e. sunitinib, sorafenib, erlotinib, gefitinib).

Agents that act on endothelial cell functions can be divided into those that (1) inhibit endothelial cell proliferation such as fumagillin analogs, squalamine and endoge- nous inhibitors endostatin and angiostatin; (2) inhibit endothelial cell invasion and motility like the matrix metalloproteinase (MMP) inhibitors (3) inhibit endothelial cell adhesion such as drugs targeting αvβ3 integrin. Other agents, often with multiple or unknown mechanisms are being investigated such as thalidomide and its analogs.

The impact anti-angiogenic agents will have on treating cancer remains unclear. As monotherapy, anti-angiogenic agents have low objective response rates [13, 14], potentially due to development of resistance by the induction of secondary pathways of angiogenesis [15]. Angiogenesis inhibitors in combination with other cytotoxic modalities may yield the best results for cancer patients [16]. Several clinical trials of angiogenesis inhibitors in conjunction with radiation therapy are underway (www.clinicaltrials.gov;[5])

Radiation and angiogenesis are connected at the molecular level [17, 18]. Cells in a hypoxic environment are resistant to radiation ([1921]. Tumors respond to radiation and stressors like hypoxia by producing hypoxia-inducible factor (HIF)-1, a strong survival mediator which inhibits apoptosis in endothelial cells [22, 23]. Inhibition of HIF-1 activity results in long-term growth suppression in tumor xenografts [24]. It would be expected that inhibition of angiogenesis would cause an increase in HIF-1 expression and hypoxia as a result of impaired vascularization leading to radioresistant states in tumors. However, HIF-1 expression also leads to VEGF transcription and angiogenesis induction [13, 2528]. With this understanding, angiogenesis inhibition in vivo might either induce radiation resistance or sensitivity.

In fact, most studies favor increased radiosensitivity with inhibition of angiogenesis. One of the earliest observations of this phenomenon of sensitization to radiation with inhibition of angiogenesis also showed an improved oxygenation in tumors subjected to anti-angiogenic therapy [29]. Recent evidence shows some normalization of vasculature with angiogenesis inhibition and a subsequent period of increased oxygenation and susceptibility to radiation [3034]. Riesterer et al. demonstrated extended tumor growth delay and tumor-cell apoptosis with combination anti-angiogenic and radiation therapy as well as reducing the hypoxic response to radiation [35].

1.1 The pursuit of surrogate markers for angiogenesis

Traditional cytotoxic cancer therapies are typically titrated to achieve a maximum tolerated dose for a selected population. Unlike conventional cytotoxic chemotherapies, targeted therapies, such as angiogenesis inhibitors, may achieve therapeutic levels long before toxicities arise [36]. For this reason, it is necessary to identify biomarkers that accurately reflect the effect of a drug on its known targets and predict response to treatment [3739]. Current angiogenesis inhibitors are typically cytostatic, and are thought to alter vessel structure instead of resulting in direct tumor kill. For this reason, investigators are aggressively pursuing suitable markers of anti-angiogenic modulation of tumor vasculature. This search has been challenging due to variations of tumor vasculature between tumor types, tumor histologies, tumor size, and degree of differentiation [4044].

In general, markers of angiogenesis inhibitor effect can be divided into three major categories: invasive measures, minimally invasive measures, and non-invasive measures. Many technologies can be applied to more than one type of biospecimen such that they can be used as an invasive (biopsy) or minimally invasive measure (serum). Each of these markers can provide different information regarding the effect of the agent on tumor vasculature. Information can vary from anatomic to physiologic. It is not yet clear whether one marker may be most appropriate in certain clinical situations or whether a panel of markers may be required for an optimal assessment of angiogenic state of the tumor. We provide a general overview of each of these classes of markers and a brief discussion of the results and limitations of several markers that have been tested in this setting (Table 3).

Table 3.

Summary of candidate markers reviewed

Marker/
method
Examples of methods
described
Invasiveness Advantages Disadvantages
MVD Immunostaining with
CD31, CD34, CD105;
 structural analysis
Invasive Potential prognostic indicator; variety of
 immunostaining methods; potential for
 structural evaluation of tumor vasculature
No clear association with other markers or AA response; invasive
 (sampling error; disruption of normal tumor biology; wound healing
 concerns with AA; ethical concerns)
Skin biopsy Visual Scoring System;
 Immunostaining
Invasive Accessible sample; no disruption of tumor
biology; reproducible physiologic process with
 continuous observation; patient is their own
 control
Invasive (wound healing concerns with AA, ethical concerns);
 non-tumor tissue may not be representative of tumor tissue
Proteomics 2D GE; Mass
Spectrometry; Protein
 Arrays; Cytometry
Invasive or
 minimally
 invasive
 applications
Ability to identify numerous proteins from
multiple sample types; applications to multiple
 aspects of cancer management
Invasive (sampling error; disruption of normal tumor biology;
 wound healing concerns with AA; ethical concerns); uncertain
 correlation of biological fluids and angiogenic state
Gene
 profiling
Microarray; SAGE Invasive or
 minimally
 invasive
 applications
Ability to identify genes from multiple sample
types; applications to multiple aspects of
 cancer management
Invasive (sampling error; disruption of normal
 tumor biology; wound healing concerns with AA;
 ethical concerns); uncertain correlation of biological fluids and angiogenic state
Growth
 factors and
 cytokines
VEGF, HGF, FGF Invasive or
 minimally
 Invasive
 applications
Ability to serially evaluate multiple aspects
 of cancer management
Inconsistent results; lack of understanding of kinetics;
 modification by other treatments;
Cell-surface
 markers
VCAM, ICAM, FLT Invasive or
 minimally
 Invasive
 applications
Probably most useful in pretreatment and
 surveillance phases
Inconsistent results; lack of understanding of kinetics;
 modification by inflammation
CECs and
 EPCs
VEGF, VE-cadherin Minimally
invasive
Ability to serially evaluate multiple aspects
 of cancer management
Inconsistent results;
 lack of understanding of kinetics; modification by other treatments;

AA Anti-angiogenic agent

2 Invasive measures: tissue biomarkers

The most intuitive method to measure the effect of any drug is to evaluate the target tissue, the tumor. Biopsies provide a way to thoroughly characterize tumor, histology and molecular processes with techniques such as immunohistochemistry, microarray, and proteomic analysis. These methods may be helpful in examining therapeutic effects of radiation, chemotherapy, targeted therapies, and their combinations. While evaluation of tissue provides an excellent mechanism to evaluate drug effect, the method raises practical and ethical concerns. The ethical implications of subjecting patients to serial biopsies in the context of a clinical trial have been debated [45, 46]. At this time, there is no consensus in the research community regarding the appropriateness of repeated biopsies.

In addition to ethical concerns, the logistical and monetary costs of multiple biopsies are significant, making this option impractical for larger studies. Smaller series have successfully used this technique and gained a wealth of information. An excellent example of the successful use of tissue biopsies to evaluate tissue effects of an anti-angiogenic agent is a study of combined chemoradiotherapy and bevacizumab in rectal cancer patients [47, 48]. Finally, the use of tissue markers requires biopsy of a portion of tumor. This technique may lead to sampling error, disruption of normal tumor biology after each biopsy, and the potential for wound healing problems when performed concurrently with the delivery of an anti-angiogenic agent, radiation, or cytotoxic chemotherapy.

2.1 Microvessel density and structure

Evaluation of microvessel density (MVD) is performed by immunostaining endothelial cells in tissue, identifying “hot spots” of angiogenesis and counting the number of vessels per high power field. Within tumors, MVD has been identified as a potential prognostic indicator of progression, overall survival, and disease-free survival in multiple histologies [4952]. MVD has also been explored as a method to predict and evaluate the efficacy of anti-angiogenic therapy [53, 54]. Regardless of the prognostic value of MVD, its utility in evaluating response to anti-angiogenic therapy has been disappointing.

Pre-clinical data [53, 55] and numerous clinical trials show that microvessel density does not predict response to anti-angiogenic therapy, nor does it predict the dose required to elicit an anti-angiogenic response [5661]. In addition, MVD does not appear to be associated with other non-invasive serum markers and imaging techniques used as markers of angiogenic state [62]. However, two recent trials in gastric and head and neck cancer patients found statistically significant decreases in MVD before and after COX-2 inhibitor therapy, which is thought to act partially as an anti-angiogenic agent [63, 64].

There are several potential reasons for the conflicting reports of the utility of MVD in the setting of evaluation of efficacy of anti-angiogenic therapy [65, 66]. For one, the choice of immunostained antigen may affect MVD prognostic and predictive value. Several studies have measured MVD with antibodies to CD105, a protein expressed in higher quantities in proliferating tumor endothelial cells compared to normal microvasculature [6770]. Because other antigens such as CD31 and CD34 are present on the surface of tumor endothelial cells regardless of proliferation status, they may not reflect the presence of targeted proliferating neo-vessels as effectively as CD105 [71].

One major concern with any biopsy marker, such as MVD, involves sampling error. As mentioned, measurement relies on selection of “hot spots” within tissue that could vary substantially. Also, the MVD of the biopsy may not be representative of the remainder of the tumor. Additionally, as MVD is a measure of vessels per area of tumor, the measurement reflects the balance of tumor cells and vessels. If a proportional number of tumor cells and vessels are eliminated with a therapy, the vessel density measurement may remain stable, even though both tumor and endothelial cells have been killed [53]. Questions also arise concerning whether MVD reflects the functionality of vasculature present as well as thedegreeofthe tumor’s dependence upon the vasculature identified [65].

Yet another concern is the affect additional treatments, besides anti-angiogenic therapy, will have on MVD. In 1945 it was discovered that radiation has an independent effect on MVD as well as vessel length and diameter. Both single dose [72, 73] and fractionated radiation [74] result in altered tumor vascularization, noted as early as 12 h post-irradiation. These changes reflect a decrease in the intercapillary distance in irradiated tumors, possibly leading to reoxygenation following radiation [74].

The concerns about MVD sampling challenges, modulation by other therapies and the observed lack of correlation with anti-angiogenic response indicate that MVD cannot be supported as a direct measure of angiogenesis in clinical trials. More promising results might be found with greater characterization of neoangiogenesis as well as more qualitative structural analysis of tumor vasculature [71]. While anti-angiogenic therapy may not consistently alter MVD, evidence suggests that microvascular structure may be “normalized” as angiogenesis is inhibited [30, 31, 33, 34, 75]. This parameter can be measured through techniques such as vascular casting or vascular contrast, which typically require large samples of tissue for evaluation. Evaluation of vessel structure during a lead-in phase with neo-adjuvant delivery of the angiogenesis inhibitor alone may help distinguish what effects are attributable to the angiogenesis inhibitor.

2.2 Proteomic analysis

The field of proteomics was developed to allow the simultaneous evaluation of numerous peptide biomarkers. The concept evolved from the theory that the pattern of expression of a number of peptides in biologic samples might provide a better diagnostic tool than the evaluation of single proteins [7678]. Advances in the field of proteomics such as 2D gel electrophoresis, mass spectrometry technologies, and protein array technologies such as surface-enhanced laser-desorption/ionization time-of-flight (SELDI-TOF) mass spectroscopy allow the generation of a descriptive “fingerprint” of polypeptide expression in serum samples [76]. The spectra generated through this analysis can be compared to the spectra of other subjects to generate a pattern predictive of the presence of cancer [7982], disease stage [83, 84], and therapeutic effect [78, 85].

Direct tissue profiling of small biopsy sections can yield full-scale proteomic analysis revealing relevant biomarkers or patterns [86, 87]. In tissue samples, techniques such as laser capture microdissection can allow collection of specific cell subtypes. This powerful technology may allow the generation of specific expression profiles from primary tumor cells, metastatic tumor cells, and endothelial cells that will allow assessment of response to each cell type to therapy [88, 89]. Additionally, by creating a fingerprint for each cell type and how they are altered in response to therapy, intermittent evaluation of serum profiles may allow an assessment of response in each cell line through shedding into the vascular compartment. The continued introduction of numerous technological advancements such as artificial intelligence-based pattern recognition algorithms will allow a more rapid and sensitive detection of patterns that will assist in detecting and monitoring cancers [78, 90, 91]. New methods like linear ion trap quadrupole mass spectrometers enhance quantification abilities [92, 93] and capillary electrophoresis mass spectrometry is allowing better evaluation of large data sets, more precise differentiation of proteins and more rapid monitoring of data quality [94].

Recent studies have investigated the use of proteomic techniques to identify proteins associated with tumor endothelial cells [9599]. The study that to date best represents this application defined 15 proteins differentially upregulated in tumor endothelium of lung metastases from breast primaries compared to normal rat lung [100]. Another study noted differential expression of four proteins in glioma vasculature not found in normal brain tissue [101]. Some of these proteins have been validated using specific antibodies to label tumor and normal tissue [100102].

Since proteomic profiles can be generated from tissue as well as biological fluids, various limitations may apply. With tissue samples, the same concerns that exist for MVD often apply here, namely sampling error, invasiveness of the procedure, and the requirement for multiple biopsies to compare prior to and after therapy. The use of this technique for evaluation of biological fluids minimizes these concerns, however it remains unclear if the use of serum proteomic profiling will allow an accurate estimation of angiogenic state within a tumor. Furthermore, it is unclear if a signature or pattern representative of altered angiogenic state will apply across multiple primary sites (variable leak into the plasma compartment), histologies, grades of tumor, and different total tumor burdens.

2.3 Gene expression profiling

Technologies such as DNA microarrays and serial analysis of gene expression (SAGE) are creating opportunities to investigate gene expression in tumors [103]. This implies the potential to use tumor biopsies, circulating tumor cells, circulating endothelial cells, and whole blood to identify new surrogate markers of angiogenesis. Multiple genes have been implicated in angiogenesis by gene expression profiling for a number of tumors [77]. St Croix et al. purified and concentrated colon cancer endothelial cells and used SAGE to identify 79 genes that were upregulated or downregulated. Among those discovered were novel tumor endothelial markers found to be overexpressed in other tumors [104, 105], some with potential therapeutic implications [106, 107]. SAGE has also been employed to identify genes in brain tumors and breast cancers [108110]. Some investigators have proposed the use of gene expression databases using similarities of ordered gene lists approach for comparisons and pooling of data to increase identification of genes involved in angiogenesis or the response to these agents [76, 111, 112]. However, the large scale combination of these data sets would be limited by technical and statistical problems.

In addition to identification of genes involved in angiogenesis, expression profiling also provides other potential applications to the investigation of angiogenesis inhibitors. With the use of dynamic contrast imaging as a guide, areas of tumors thought to be active from an angiogenic standpoint can be targeted for biopsy [113115]. Techniques such as laser capture microdissection allow comparisons of expression profiles in regions of tumors thought to be of interest [113, 116119]. By evaluating which proteins are expressed in regions of tumors with significant angiogenic activity, tumors from protocol candidates may be screened for expression to help determine the relative angiogenic state of their tumor. Once gene expression profiles of tumors responsive to angiogenic therapy or combination angiogenic and cytotoxic therapy are known, an initial tissue biopsy could help determine which patients would benefit prior to initiation of therapy. Finally, by evaluating changes in tumors which are a result of angiogenic therapy or the combination of angiogenic therapy and radiation, the mechanisms of the additive and synergistic responses may be better understood. These techniques require the ability to safely and accurately target a specific region of a tumor based on imaging.

2.4 Skin biopsies

Angiogenesis is known to play an important role in wound healing, and the effect of anti-angiogenic treatment on wound healing has been studied in clinical trials [120]. Because angiogenesis is required for wound healing, skin biopsies may be helpful in evaluating the response to anti-angiogenic agents or in titrating agents to an appropriate dose by performing serial punch biopsies of the skin, each time removing the previously biopsied site (Reviewed in [121]). Clearly, the same ethical issues for multiple biopsies apply in this instance as well. In addition, it is not entirely clear that inhibition of angiogenesis in a well-vascularized skin wound correlates with that observed in the heterogenous environment of a tumor. Few studies have performed this technique.

Zhang et al. evaluated anti-angiogenic effects of MEDI-522, a monoclonal antibody to αvβ3 integrin, in a phase I dose-escalation study. Pre-treatment and post-treatment punch biopsies of the skin were assessed for vascular area, endothelial cell proliferation and apoptosis, and β3 integrin levels. None of the above parameters after MEDI-522 treatment were found to be significantly different when compared to the pre-treatment tissue in spite of adequate drug presence identified in the vasculature by immunohistochemistry [122]. Mundhenke et al. evaluated the response to endostatin in patients by serial skin biopsies. While no significant changes were noted in vascular density or blood vessel maturity in biopsied skin, it is important to note that changes were also not appreciated in tumor biopsies after endostatin therapy [123]. Lockhart et al. used a similar approach to assess wound healing and the activity of a matrix metalloproteinase inhibitor. Rather than focusing on specific angiogenic parameters, they recorded wound healing by visual wound assessment, reporting a statistically significant difference in time to target healing level between treatment and control groups [124].

These studies show that this method is practical and well-tolerated. With continued investigation, this innovative procedure or similarly designed methods might play a role in assessment of individual response to anti-angiogenic drugs at various dose levels.

3 Minimally invasive measures: circulating markers

Identifying circulating markers of angiogenesis that could assist in diagnosis, staging, treatment response and follow-up is being aggressively pursued. Such markers would have the advantage of being minimally invasive allowing repetitive sampling throughout treatment and follow-up without the ethical and technical implications of multiple biopsies. In addition, sampling for these minimally invasive markers would not disrupt tumor physiology. There are currently three main categories of markers being investigated as minimally invasive measures of angiogenic state including growth factors and cytokines, cell surface molecules, and circulating endothelial and endothelial precursor cells.

3.1 Growth factors and cytokines

Multiple angiogenesis growth factors and cytokines in blood and urine have been investigated in a range of tumor histologies. For example, elevated VEGF, FGF, and HGF in various biological fluids have been associated with staging, progression, and prognosis [125139]. Similar to most markers of angiogenesis, studies in this area have yielded conflicting results [140142]. Various factors may reflect the varying results obtained with these markers, including microenvironmental variations such as vascular permeability altering the quantity of protein that intravasates into the blood stream [65]. Another possible confounding factor includes tumor heterogeneity in regards to elaboration of these factors.

Recent studies have shown promising results for use of growth factors as a marker of anti-angiogenic response in a variety of treatment conditions ([143, 144]. Drevs et al. described a time- and dose- dependent reduction in soluble VEGFR-2 with once-daily oral AZD2171 in 36 patients with solid tumors and liver metastases. Increases in VEGF and PlGF were detected after treatment, but there was no suggestion of a dose relationship [145]. This increase in VEGF after anti-angiogenic treatment has been reported often since first noted in 2003 [14, 146].

Because most anti-angiogenic agents are combined with other modalities, it is important to understand how other therapies perturb the levels of these growth factors as well. Similar correlations have also been reported in anti-angiogenic therapies combined with other treatments [147]. Evaluations of these growth factors with radiation have yielded interesting results, with a clear time dependence of the kinetics of these markers after therapy and a correlation with outcome. Chan et al. found significant predictive value in the kinetics of urine VEGF levels in patients treated with one month of radiation for various tumor types. Increasing serum levels of VEGF after radiation were correlated with eventual failure or progression [127]. Ria et al. found decreases in serum FGF-2, VEGF, and HGF following radiation to primary or metastatic tumors of various histologies, with a correlation between radiation dose and decreases in serum FGF-2 and VEGF [148]. Others have found the higher pretreatment serum VEGF levels to be predictive of poor outcome following combined chemotherapy and radiation [149, 150].

More data regarding the kinetics and utility of these minimally invasive markers are needed to successfully incorporate them into clinical use. Many ongoing trials continue to include these important markers, hopefully leading to firmer conclusions about their utility in diagnosing, staging and following patients treated with standard and anti-angiogenic therapies.

3.2 Endothelial cell surface molecules

Endothelial cells release various molecules into the circulation, implying the potential for angiogenesis markers related to cellular adhesion. Some soluble molecules (i.e. sVCAM-1, sICAM-1, sFLT-1) have been elevated in patients with cancers, relative to normal controls or patients with benign neoplasms [151156]. Shariat et al. found that circulating sVCAM-1 levels increased incrementally from healthy controls to prostate cancer patients with localized disease and then to those with lymphatic spread and metastasis. In a pre-operative model, elevated plasma sVCAM-1 and VEGF were both associated with biochemical progression [136]. Since inflammatory processes in the vasculature may cause elevation of cell surface markers, inflammatory response to radiation therapy may alter the kinetics of these molecules [157, 158]. As with other potential markers, cell surface markers for angiogenesis may be more useful in the pretreatment and surveillance setting due to perturbations during therapy by other concurrent therapies such as cytotoxic chemotherapy and radiotherapy.

3.3 Circulating endothelial and endothelial precursor cells

Tumor angiogenesis involves locally derived circulating endothelial cells (CECs) and bone marrow-derived endothelial precursor cells (EPCs) [159]. As tumors grow, pro-angiogenic molecules recruit nearby tumor endothelial cells, perivascular cells, and circulating EPCs to the vascular bed [160, 161]. Anti-angiogenic agents have been shown to inhibit EPC mobilization. [162]. A sufficiently strong correlation between CECs and EPCs and angiogenesis appears to support the potential use of these cells to monitor anti-angiogenic effects [163].

Measurement of CECs and EPCs has been reported in vascular surgery patients and following myocardial infarction [164, 165]. Recently, Norden et al. treated imitinab-resistant gastrointestinal stromal tumor patients with sunitinib. In this series, the presence of VEGF bearing CECs and monocytes differentiated patients with progressive disease and those who exhibited a clinical response [144]. Another potential marker from endothelial cells is VE-Cadherin. One study measured elevated circulating VE-Cadherin RNA levels in breast cancer patients and pregnant women but found none in healthy controls [166]. As VE-Cadherin is an endothelial-specific gene, it is proposed that this may be a marker for CECs. Measuring CECs and EPCs in cancer patients undergoing anti-angiogenic therapy may allow titration of dose to the desired effect. However, before these markers will be effectively incorporated into clinical use, further characterization of these cells’ response to other modalities is needed to understand the complex interactions which appear to be present. For example, Furstenberger et al. described an elevation of CECs and EPCs in 10 patients with locally advanced breast cancer. Neoadjuvant chemotherapy resulted in a decrease in CECs and an increase in EPCs [167].

4 Non-invasive markers: imaging

The application of existing imaging technologies to the measurement of metabolism, oxygenation, and perfusion is a field of intense research (Table 4) [62, 121, 168170]. Imaging offers the distinct advantage of being able to serially evaluate anatomical and physiological processes in tumors without disrupting tumor tissue. To use these non-invasive methods, a better understanding of the invasive correlates of the images and information obtained with these technologies is needed. In addition, if the various imaging modalities available are to be effectively incorporated into widespread clinical trials, standardization is necessary.

Table 4.

Summary of imaging reviewed

Modality Examples of specific methods described Advantages Disadvantages
MRI DCE-MRI, antibody contrast agents (αvβ3
 integrin, E-selectin)
Excellent anatomic information and spatial resolution; highly
 sensitive; multiple types of low-toxicity contrast; no radiation
 exposure
Expensive; motion effects; lack of targeted probes; longer
 procedure time
CT Functional CT Excellent anatomical information and spatial resolution; simple
 quantitation; direct proportion of contrast
Recurrent ionizing radiation exposure; contrast agent toxicity;
 lack of targeted probes
PET FDG, [18]F-FMISO, [15]O H2O, [15]O
 CO, radiolabeled peptides
Whole body imaging; direct proportion to contrast; high
 sensitivity; high throughput; ability to use multiple tracers
Expensive with limited availability; limited spatial resolution;
 lack of anatomical information; short radionuclide half-life
limited penetration; interoperator variability; lack of targeted
 probes
Ultrasound Color Doppler; power Doppler;
 microbubbles; antibody contrast agents
 (αvβ3 integrin)
Sensitive applications; anatomical imaging; inexpensive and
 widely available; portable;
Optical
 imaging
Near infra-red Highly sensitive; inexpensive; portable Poor tissue penetration allowing for superficial sites only; lack
 of anatomical information; technology still in early
 development

4.1 PET

Positron emission tomography (PET) has played an ever increasing role in the staging of malignancy and assessment of response to anticancer therapy. The use of fluoro-deoxy-D-glucose (FDG) PET to evaluate the response to anti-angiogenic therapy or radiation has been described in variety of tumor types and disease sites [121, 171175]. This is complicated by the fact that hypoxia, theoretically induced by anti-angiogenic agents, increases the expression of the Glut-1 glucose transporter and the uptake of FDG into tumor cells ([176, 177]. Other variables, such as the proportion of metabolism due to aerobic versus anaerobic mechanisms may also affect the accumulation of FDG [178]. The effects of the tumor microenvironment on FDG uptake are incompletely understood and complicate the quantitation of FDG accumulation and comparisons between tumors in different subjects.

While FDG PET plays an important role in modern oncology, the development of new tracers for PET imaging promises to improve the understanding of the physiologic state of individual tumors. Physiologic variables such as tumor blood flow, metabolism, and hypoxia can be measured with PET utilizing various specialized tracers. For example, tumor hypoxia can be imaged by utilizing nitroimidazole tracers that are absorbed into tissues and trapped in their reduced state [179183]. Cher et al. showed that 18F-FMISO PET provided a noninvasive assessment of hypoxia in glioma that was prognostic for treatment outcomes in the majority of patients, and hypothesized that it may have a future role in monitoring anti-angiogenic treatment [184]. The use of 18F-FMISO PET has been incorporated into clinical trials of chemoradiation in conjunction with tirapazamine, a toxin targeting hypoxic cells [185187] To our knowledge, this has not yet been included in human clinical trials with anti-angiogenic agents.

Tumor blood flow can be measured with the use of H152O as a tracer. As water is freely diffusible, this tracer will rapidly equilibrate and is metabolically inert. A number of laboratory studies have assessed this technique in combination with anti-angiogenic agents [181, 188, 189]. Clinical studies with radiation, chemotherapy or anti-angiogenic agents have incorporated this technique as a measure of tumor perfusion [62, 171, 174, 190193]. It remains to be determined whether H152O PET imaging will play a significant role in assessing response to anti-angiogenic agents in the clinic. Another tracer developed to measure vascular volume is C15O which binds to the hemoglobin of red blood cells. A combination of H152O and C15O may be the best method to reflect vessel density changes [194].

Design and manufacture of PET probes that bind to signaling intermediates or are ligands for angiogenic receptors are under investigation and may help to better define the angiogenic state of tumors [195203]. Examples include radiolabeled peptides that bind to VEGFR [204, 205] and also to the αvβ3 integrin receptor expressed on endothelial cells [206, 207]. These approaches are being actively evaluated in the laboratory, and it is unclear what role they will play in the clinic.

Limitations of PET imaging include the relatively poor anatomic resolution compared to CT and MRI and the requirement of a radioactive isotope generated in a cyclotron. Additionally, attempts to quantify PET data require an ability to correct for the attenuation of emitted photons in tissue [181, 208]. Registration of PET images to computed tomography (CT) or magnetic resonance imaging (MRI) can significantly improve the diagnostic accuracy of PET [209, 210], partially correcting for decreased anatomic resolution of PET by integrating the anatomic data of CT.

4.2 MRI

Dynamic contrast-enhanced MRI (DCE-MRI) is currently the most useful method for assessing early changes in tumor vasculature in clinical trials [211, 212]. The technique involves the acquisition of magnetic resonance images before, during, and after the delivery of contrast to evaluate physiologic parameters such as perfusion and capillary permeability [213]. DCE-MRI can distinguish malignant and benign tissue based on differences in the function of tumor microvasculature [214]. A number of contrast agents can be used to perform DCE-MRI, including low molecular weight agents, high molecular weight agents, and agents that accumulate at sites of angiogenesis (reviewed in [168, 215]. The choice of contrast agents for DCE-MRI depends on the physiologic process to be evaluated [211].

Low molecular weight agents rapidly diffuse into the extracellular fluid space, with as much as 12–45% of the contrast media passing into the extracellular space during the first pass [216]. Tissue perfusion and blood volume can be measured using T1 and T2-weighted MR imaging sequences with low molecular weight contrast enhancement [217]. In addition, regions of tumors with necrosis and fibrosis have differing vascular function which can be visualized with these techniques [214].

Tumor vasculature is highly permeable to macromolecules including high molecular weight contrast agents [218]. These agents can be used to assess changes in vascular permeability in tumors after treatment with radiation [201] or anti-angiogenic therapy.

Agents designed to image tumors by binding to angiogenic molecules have been evaluated in preclinical studies including ανβ3 integrin and E-selectin antibody contrast agents [122, 219223]. These agents may be difficult to image due to the low concentration of their target in tumors, typically below the detection level for MRI [224]. Solutions to this problem would include agents designed to accumulate at sights of angiogenesis through modulation or amplification [170].

Some difficulties in the use of DCE-MRI to assess angiogenesis have become apparent. Studies evaluating the correlation of microvessel density and DCE-MRI have found conflicting results [213, 225]. This discrepancy may be due to a lack of correlation between vessel permeability and perfusion. Some regions of the body are better candidates for evaluation with DCE-MRI due to technical considerations such as immobilization. Registration of images can be difficult for non-stationary organs, and lack of immobilization resulting in motion artifact may cause difficulties in image sequence registration [226]. Additional concerns for DCE-MRI imaging include the injection rate and the timing of sequence timing [213].

Quantitation of data obtained with DCE-MRI is complex and requires consideration of multiple variables corrected with standardized values taking into account the patient’s weight and cardiac output. Technical limitations and physiologic considerations that affect the accuracy of quantitation with DCE-MRI have been described in detail [211, 214].

Anti-angiogenic agents have been shown to reduce tumor vascular permeability in pre-clinical studies [227, 228]. Several clinical trials evaluating alterations in DCE-MRI parameters following therapy with anti-angiogenic agents have been completed with promising results [61, 229232]. Liu et al. showed a decrease in vascular parameters measured by DCE-MRI after anti-angiogenic therapy that indicate utility as an indicator of drug pharmacokinetics [233]. However, questions remain about the optimal timing of DCE-MRI evaluation in relation to delivery of anti-angiogenic drugs [214] in order to appropriately evaluate for response. Regardless, recent clinical trials have demonstrated that measurements obtained from DCE-MRI correlate with plasma concentration of anti-angiogenic agents and can potentially predict clinical response after anti-angiogenic treatment [61, 234236].

4.3 CT

Functional CT imaging may provide an evaluation of tumor blood flow, blood volume, and permeability [237239]. Many of the concepts used to evaluate these variables with MRI can be extended to CT. The procedure involves injection of a contrast agent followed by serial evaluations at various time points to assess these physiologic endpoints ([65]. Advantages of CT include a linear relationship between signal and contrast concentration allowing for simple quantification and widespread availability. However, lack of experience and technology with CT and concerns about recurrent exposure to ionizing radiation have limited its progress as a marker.

The procedure has been used to evaluate the angiogenic state of tumors and compared favorably to MVD [240242]. Ma et al. found that sixteen-slice spiral CT perfusion imaging was significantly associated with tumor angiogenesis and reflected MVD measurement and cyclin D1 expression in untreated peripheral lung cancer patients less than 1 week before surgery [243]. Other trials have investigated angiogenic evaluation with functional CT for different malignancies before and after anti-angiogenic therapy with both mixed results [244247]. Comparisons of contrast-enhanced dynamic CT and DCE-MRI show similar results can be obtained with either modality [248].

4.4 Ultrasound

Ultrasound is playing an ever increasing role in the staging of primary tumors and metastatic disease in a variety of malignancies including lung, gastrointestinal, and urologic malignancies [249254]. The ability to apply ultrasound probes in proximity to these tumors in concert with numerous technical advancements allows accurate determination of tumor depth and lymph node involvement.

In addition to anatomic imaging, ultrasound can evaluate tumor blood flow and the tumor microvasculature. Advancements such as color Doppler and power Doppler allow assessment of response to anticancer therapy with an assessment of tumor blood flow and microvascular anatomy [255]. Color Doppler imaging allows quantitation of blood flow through computerized image analysis [256]. Unfortunately, the low velocity of capillary flow is typically not evaluable by Doppler sonography [257]. The use of blood pool ultrasound contrast agents has allowed the investigation of smaller vessels, including those measuring 30 to 60 μm[170].

Ultrasound has also been evaluated for molecular imaging using ultrasound contrast agents designed to bind to specific ligands such as the ανβ3 integrin [258]. Analysis of the amount of bound targeted ultrasound contrast through ultrasound induced microbubble destruction may allow quantitation of contrast binding [170] and can demonstrate vessels as small as 70 μm[121]. Advances such as these may allow an evaluation of the density or function of specific receptors in tumors that an anti-angiogenic therapy may target.

Ultrasound is being used increasingly in clinical trials to evaluate the vasculature in tumors, and seems to compare favorably with more established techniques [259, 260]. Transcranial imaging has been shown to be effective for evaluating blood flow in high grade gliomas, with similar results as those obtained with perfusion MR imaging [259]. Ultrasound has effectively been used to evaluate the response of preclinical tumor models and human tumors to anti-angiogenic drugs [261263]. As with any marker, conflicting results have been obtained with attempts at clinical translation. In one Phase I trial, ultrasound blood flow parameters in metastatic liver tumors were explored as a marker for PTK/ZK response. Only a non-statistically significant trend towards higher blood flow with increasing doses of drug was seen and no dose-related changes were noted for a calculated resistance index [264].

While ultrasound may provide an accurate prediction of blood flow, a major problem with the incorporation of Doppler imaging into a clinical trial is the dependence on experienced operators [265]. A lack of experience may lead to significant inter- and intra-observer variability in measurements, complicating longitudinal evaluations. Additionally, physical characteristics of the tissues through which tumors will be visualized can affect the quality of imaging. Benefits to this technology include the relatively low cost, portability, and non-invasive nature of the procedure [255].

4.5 Optical imaging

Technologies such as near-infrared spectroscopic diffuse tomography and orthogonal polarization spectroscopy are under evaluation for their utility in imaging angiogenic vasculature. Optical imaging generates images using measurements of visible or near-infrared light scattered across human tissues [266]. The technology is inexpensive and portable, but the consistent challenge has remained the limited penetration and intense scattering of light. Nevertheless, optical imaging is considered feasible in superficial tumors such as in the breast, eye and pediatric neoplasms [267]. Some trials investigating the sensitivity and specificity of optical imaging techniques measuring total hemoglobin and relative oxygenation to compare normal to malignant breast tissue [268] and to differentiate malignant from benign breast masses [269] have been performed with promising results [270].

5 Conclusion

Anti-angiogenic therapies are part of a growing body of molecularly targeted therapies for cancer. Such treatments are imposing changes on the process of drug development, evaluation and approval. Recent interpretations of FDA regulations allow for phase 0 clinical trials to be performed, which would involve developing assays to evaluate target modulation and tissue effects of a drug and to obtain preliminary pharmacokinetic data [36]. The benefits of this process cannot be realized without the use of accurate biomarkers.

Numerous candidate markers of angiogenesis have been identified, but the use of these markers in diagnosis, prognosis, and monitoring of treatment remains investigational and of uncertain utility. It is improbable that any one biomarker will provide all relevant clinical information in the setting of a trial of anti-angiogenic therapy alone or in combination with additional cytotoxic therapies. Rather, a combination of markers obtained from tissue, biological fluids, and imaging is more likely to result in a comprehensive understanding of the complex process of angiogenesis and any perturbations from therapy. Additionally, putative markers will probably vary with differences in tumor attributes (histology, size, proliferation rate, etc) and the treatment regimen employed. Future clinical trials of anti-angiogenic agents should seek to forward discovery of new biomarkers and to validate promising candidate markers and imaging modalities already described. Following this pattern in the progress of anti-angiogenic therapy will hopefully lead to better outcomes in cancer patients while establishing a model for investigation of future molecularly targeted therapies.

Acknowledgements

AB: This research was supported by the Intramural Research Program of the NIH, NCI. This research year was made possible through the Clinical Research Training Program, a public—private partnership supported jointly by the NIH and Pfizer Inc (via a grant to the Foundation for NIH from Pfizer Inc). DC, KC: This research was supported by the Intramural Research Program of the NIH, NCI.

References

  • 1.Folkman J. Tumor angiogenesis: Therapeutic implications. The New England Journal of Medicine. 1971;285(21):1182–1186. doi: 10.1056/NEJM197111182852108. [DOI] [PubMed] [Google Scholar]
  • 2.Carmeliet P. Angiogenesis in life, disease and medicine. Nature. 2005;438(7070):932–936. doi: 10.1038/nature04478. [DOI] [PubMed] [Google Scholar]
  • 3.Carmeliet P, Jain RK. Angiogenesis in cancer and other diseases. Nature. 2000;407(6801):249–257. doi: 10.1038/35025220. [DOI] [PubMed] [Google Scholar]
  • 4.Ferrara N, Kerbel RS. Angiogenesis as a therapeutic target. Nature. 2005;438(7070):967–974. doi: 10.1038/nature04483. [DOI] [PubMed] [Google Scholar]
  • 5.Nieder C, et al. Current status of angiogenesis inhibitors combined with radiation therapy. Cancer Treatment Reviews. 2006;32(5):348–364. doi: 10.1016/j.ctrv.2006.03.006. [DOI] [PubMed] [Google Scholar]
  • 6.Quesada AR, Munoz-Chapuli R, Medina MA. Anti-angiogenic drugs: From bench to clinical trials. Medicinal Research Reviews. 2006;26(4):483–530. doi: 10.1002/med.20059. [DOI] [PubMed] [Google Scholar]
  • 7.Walsh DA. Pathophysiological mechanisms of angio-genesis. Advances in Clinical Chemistry. 2007;44:187–221. doi: 10.1016/s0065-2423(07)44006-9. [DOI] [PubMed] [Google Scholar]
  • 8.Ferrara N. Vascular endothelial growth factor: Basic science and clinical progress. Endocrine Reviews. 2004;25(4):581–611. doi: 10.1210/er.2003-0027. [DOI] [PubMed] [Google Scholar]
  • 9.Longo R, Gasparini G. Challenges for patient selection with VEGF inhibitors. Cancer Chemotherapy and Pharmacology. 2007;60(2):151–170. doi: 10.1007/s00280-006-0403-6. [DOI] [PubMed] [Google Scholar]
  • 10.Moreira IS, Fernandes PA, Ramos MJ. Vascular endothelial growth factor (VEGF) inhibition—A critical review. Anti-Cancer Agents in Medicinal Chemistry. 2007;7(2):223–245. doi: 10.2174/187152007780058687. [DOI] [PubMed] [Google Scholar]
  • 11.Arora N, et al. Vascular endothelial growth factor chimeric toxin is highly active against endothelial cells. Cancer Research. 1999;59(1):183–188. [PubMed] [Google Scholar]
  • 12.Frankel AE. Increased sophistication of immunotoxins. Clinical Cancer Research. 2002;8(4):942–944. [PubMed] [Google Scholar]
  • 13.Hicklin DJ, Ellis LM. Role of the vascular endothelial growth factor pathway in tumor growth and angiogenesis. Journal of Clinical Oncology. 2005;23(5):1011–1027. doi: 10.1200/JCO.2005.06.081. [DOI] [PubMed] [Google Scholar]
  • 14.Yang JC, et al. A randomized trial of bevacizumab, an anti-vascular endothelial growth factor antibody, for metastatic renal cancer. The New England Journal of Medicine. 2003;349(5):427–434. doi: 10.1056/NEJMoa021491. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Moffat BA, et al. Inhibition of vascular endothelial growth factor (VEGF)-A causes a paradoxical increase in tumor blood flow and up-regulation of VEGF-D. Clinical Cancer Research. 2006;12(5):1525–1532. doi: 10.1158/1078-0432.CCR-05-1408. [DOI] [PubMed] [Google Scholar]
  • 16.Senan S, Smit EF. Design of clinical trials of radiation combined with antiangiogenic therapy. Oncologist. 2007;12(4):465–477. doi: 10.1634/theoncologist.12-4-465. [DOI] [PubMed] [Google Scholar]
  • 17.Folkman J, Camphausen K. CANCER: Enhanced: What does radiotherapy do to endothelial cells? Science. 2001;293(5528):227–228. doi: 10.1126/science.1062892. [DOI] [PubMed] [Google Scholar]
  • 18.Wachsberger P, Burd R, Dicker AP. Tumor response to ionizing radiation combined with antiangiogenesis or vascular targeting agents: Exploring mechanisms of interaction. Clinical Cancer Research. 2003;9(6):1957–1971. [PubMed] [Google Scholar]
  • 19.Brizel DM, et al. Oxygenation of head and neck cancer: Changes during radiotherapy and impact on treatment outcome. Radiotherapy and Oncology. 1999;53(2):113–117. doi: 10.1016/s0167-8140(99)00102-4. [DOI] [PubMed] [Google Scholar]
  • 20.Brizel DM, et al. Tumor oxygenation predicts for the likelihood of distant metastases in human soft tissue sarcoma. Cancer Research. 1996;56(5):941–943. [PubMed] [Google Scholar]
  • 21.Hall E. Radiobiology for the radiologist. 5th ed Lippincott, Williams, & Wilkins; Philadelphia: 2000. [Google Scholar]
  • 22.Dewhirst MW, et al. Exploring the role of HIF-1 in early angiogenesis and response to radiotherapy. Radiotherapy and Oncology. 2007;83(3):249–255. doi: 10.1016/j.radonc.2007.05.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Moeller BJ, et al. Radiation activates HIF-1 to regulate vascular radiosensitivity in tumors: Role of reoxygenation, free radicals, and stress granules. Cancer Cell. 2004;5(5):429–441. doi: 10.1016/s1535-6108(04)00115-1. [DOI] [PubMed] [Google Scholar]
  • 24.Harada H, et al. Significance of HIF-1-active cells in angiogenesis and radioresistance. Oncogene. 2007;26:7508–7516. doi: 10.1038/sj.onc.1210556. [DOI] [PubMed] [Google Scholar]
  • 25.Gaffney DK, et al. Epidermal growth factor receptor (EGFR) and vascular endothelial growth factor (VEGF) negatively affect overall survival in carcinoma of the cervix treated with radiotherapy. International Journal of Radiation Oncology, Biology, Physics. 2003;56(4):922–928. doi: 10.1016/s0360-3016(03)00209-8. [DOI] [PubMed] [Google Scholar]
  • 26.Gorski DH, et al. Blockage of the vascular endothelial growth factor stress response increases the antitumor effects of ionizing radiation. Cancer Research. 1999;59(14):3374–3378. [PubMed] [Google Scholar]
  • 27.Kermani P, et al. Effect of ionizing radiation on thymidine uptake, differentiation, and VEGFR2 receptor expression in endothelial cells: The role of VEGF(165) International Journal of Radiation Oncology, Biology, Physics. 2001;50(1):213–220. doi: 10.1016/s0360-3016(01)01445-6. [DOI] [PubMed] [Google Scholar]
  • 28.Sonveaux P, et al. Irradiation-induced angiogenesis through the up-regulation of the nitric oxide pathway: Implications for tumor radiotherapy. Cancer Research. 2003;63(5):1012–1019. [PubMed] [Google Scholar]
  • 29.Teicher BA, Sotomayor EA, Huang ZD. Antiangiogenic agents potentiate cytotoxic cancer therapies against primary and metastatic disease. Cancer Research. 1992;52(23):6702–6704. [PubMed] [Google Scholar]
  • 30.Batchelor TT, et al. AZD2171, a pan-VEGF receptor tyrosine kinase inhibitor, normalizes tumor vasculature and alleviates edema in glioblastoma patients. Cancer Cell. 2007;11(1):83–95. doi: 10.1016/j.ccr.2006.11.021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Dings RP, et al. Scheduling of radiation with angiogenesis inhibitors anginex and Avastin improves therapeutic outcome via vessel normalization. Clinical Cancer Research. 2007;13(11):3395–3402. doi: 10.1158/1078-0432.CCR-06-2441. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Fukumura D, Jain RK. Tumor microenvironment abnormalities: Causes, consequences, and strategies to normalize. Journal of Cellular Biochemistry. 2007;101(4):937–949. doi: 10.1002/jcb.21187. [DOI] [PubMed] [Google Scholar]
  • 33.Fukumura D, Jain RK. Tumor microvasculature and microenvironment: Targets for anti-angiogenesis and normalization. Microvascular Research. 2007;74:72–84. doi: 10.1016/j.mvr.2007.05.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Winkler F, 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. 2004;6(6):553–563. doi: 10.1016/j.ccr.2004.10.011. [DOI] [PubMed] [Google Scholar]
  • 35.Riesterer O, et al. Ionizing radiation antagonizes tumor hypoxia induced by antiangiogenic treatment. Clinical Cancer Research. 2006;12(11 Pt 1):3518–3524. doi: 10.1158/1078-0432.CCR-05-2816. [DOI] [PubMed] [Google Scholar]
  • 36.Kummar S, et al. Compressing drug development timelines in oncology using phase ‘0’ trials. Nature Reviews. Cancer. 2007;7(2):131–139. doi: 10.1038/nrc2066. [DOI] [PubMed] [Google Scholar]
  • 37.Citrin D, Menard C, Camphausen K. Combining radiotherapy and angiogenesis inhibitors: Clinical trial design. International Journal of Radiation Oncology, Biology, Physics. 2006;64(1):15–25. doi: 10.1016/j.ijrobp.2005.03.065. [DOI] [PubMed] [Google Scholar]
  • 38.Jubb AM, et al. Predicting benefit from anti-angiogenic agents in malignancy. Nature Reviews. Cancer. 2006;6(8):626–635. doi: 10.1038/nrc1946. [DOI] [PubMed] [Google Scholar]
  • 39.Korn EL, et al. Clinical trial designs for cytostatic agents: Are new approaches needed? Journal of Clinical Oncology. 2001;19(1):265–272. doi: 10.1200/JCO.2001.19.1.265. [DOI] [PubMed] [Google Scholar]
  • 40.Bernsen HJ, et al. Vascularity and perfusion of human gliomas xenografted in the athymic nude mouse. British Journal of Cancer. 1995;71(4):721–726. doi: 10.1038/bjc.1995.141. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Bussink J, Kaanders JH, van der Kogel AJ. Tumor hypoxia at the micro-regional level: Clinical relevance and predictive value of exogenous and endogenous hypoxic cell markers. Radiotherapy and Oncology. 2003;67(1):3–15. doi: 10.1016/s0167-8140(03)00011-2. [DOI] [PubMed] [Google Scholar]
  • 42.Macchiarini P, et al. Relation of neovascularisation to metastasis of non-small-cell lung cancer. Lancet. 1992;340(8812):145–146. doi: 10.1016/0140-6736(92)93217-b. [DOI] [PubMed] [Google Scholar]
  • 43.Weidner N, et al. Tumor angiogenesis and metastasis-correlation in invasive breast carcinoma. The New England Journal of Medicine. 1991;324(1):1–8. doi: 10.1056/NEJM199101033240101. [DOI] [PubMed] [Google Scholar]
  • 44.Zhong H, et al. Overexpression of hypoxia-inducible factor 1alpha in common human cancers and their metastases. Cancer Research. 1999;59(22):5830–5835. [PubMed] [Google Scholar]
  • 45.Agulnik M, et al. Impact and perceptions of mandatory tumor biopsies for correlative studies in clinical trials of novel anticancer agents. Journal of Clinical Oncology. 2006;24(30):4801–4807. doi: 10.1200/JCO.2005.03.4496. [DOI] [PubMed] [Google Scholar]
  • 46.Helft PR, Daugherty CK. Are we taking without giving in return? The ethics of research-related biopsies and the benefits of clinical trial participation. Journal of Clinical Oncology. 2006;24(30):4793–4795. doi: 10.1200/JCO.2006.05.7125. [DOI] [PubMed] [Google Scholar]
  • 47.Willett CG, et al. Direct evidence that the VEGF-specific antibody bevacizumab has antivascular effects in human rectal cancer. Nature Medicine. 2004;10(2):145–147. doi: 10.1038/nm988. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Willett CG, 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. Journal of Clinical Oncology. 2005;23(31):8136–8139. doi: 10.1200/JCO.2005.02.5635. [DOI] [PubMed] [Google Scholar]
  • 49.Kolev Y, et al. Prognostic significance of VEGF expression in correlation with COX-2, microvessel density, and clinicopathological characteristics in human gastric carcinoma. Annals of Surgical Oncology. 14:2738–2747. doi: 10.1245/s10434-007-9484-7. [DOI] [PubMed] [Google Scholar]
  • 50.Lentsch EJ, et al. Microvessel density in head and neck squamous cell carcinoma primary tumors and its correlation with clinical staging parameters. Laryngoscope. 2006;116(3):397–400. doi: 10.1097/01.MLG.0000195286.29613.E1. [DOI] [PubMed] [Google Scholar]
  • 51.Nieto Y, et al. Prognostic analysis of tumour angiogenesis, determined by microvessel density and expression of vascular endothelial growth factor, in high-risk primary breast cancer patients treated with high-dose chemotherapy. British Journal of Cancer. 2007;97(3):391–397. doi: 10.1038/sj.bjc.6603875. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Uzzan B, et al. Microvessel density as a prognostic factor in women with breast cancer: a systematic review of the literature and meta-analysis. Cancer Research. 2004;64(9):2941–2955. doi: 10.1158/0008-5472.can-03-1957. [DOI] [PubMed] [Google Scholar]
  • 53.Hlatky L, Hahnfeldt P, Folkman J. Clinical application of antiangiogenic therapy: microvessel density, what it does and doesn’t tell us. Journal of the National Cancer Institute. 2002;94(12):883–893. doi: 10.1093/jnci/94.12.883. [DOI] [PubMed] [Google Scholar]
  • 54.Tozer GM. Measuring tumour vascular response to antivascular and antiangiogenic drugs. British Journal of Radiology. 2003;76(suppl1):S23–S35. doi: 10.1259/bjr/30165281. [DOI] [PubMed] [Google Scholar]
  • 55.Beecken WD, et al. Effect of antiangiogenic therapy on slowly growing, poorly vascularized tumors in mice. Journal of the National Cancer Institute. 2001;93(5):382–387. doi: 10.1093/jnci/93.5.382. [DOI] [PubMed] [Google Scholar]
  • 56.Bertolini F, Martinelli G, Goldhirsch A. Mosaic tumour blood vessels and high-dose chemotherapy for breast cancer. Lancet Oncology. 2001;2(10):595. doi: 10.1016/S1470-2045(01)00514-9. [DOI] [PubMed] [Google Scholar]
  • 57.Chhieng DC, et al. Microvessel density and vascular endothelial growth factor expression in infiltrating lobular mammary carcinoma. The Breast Journal. 2003;9(3):200–207. doi: 10.1046/j.1524-4741.2003.09311.x. [DOI] [PubMed] [Google Scholar]
  • 58.Dahut WL, et al. Phase I clinical trial of oral 2-methoxyestradiol, an antiangiogenic and apoptotic agent, in patients with solid tumors. Cancer Biology & Therapy. 2006;5(1):22–27. doi: 10.4161/cbt.5.1.2349. [DOI] [PubMed] [Google Scholar]
  • 59.Dowlati A, et al. Novel Phase I dose de-escalation design trial to determine the biological modulatory dose of the antiangiogenic agent SU5416. Clinical Cancer Research. 2005;11(21):7938–7944. doi: 10.1158/1078-0432.CCR-04-2538. [DOI] [PubMed] [Google Scholar]
  • 60.Singhal S, et al. Antitumor activity of thalidomide in refractory multiple myeloma. The New England Journal of Medicine. 1999;341(21):1565–1571. doi: 10.1056/NEJM199911183412102. [DOI] [PubMed] [Google Scholar]
  • 61.Wedam SB, et al. Antiangiogenic and antitumor effects of bevacizumab in patients with inflammatory and locally advanced breast cancer. Journal of Clinical Oncology. 2006;24(5):769–777. doi: 10.1200/JCO.2005.03.4645. [DOI] [PubMed] [Google Scholar]
  • 62.Miller JC, et al. Imaging angiogenesis: applications and potential for drug development. Journal of the National Cancer Institute. 2005;97(3):172–187. doi: 10.1093/jnci/dji023. [DOI] [PubMed] [Google Scholar]
  • 63.Soo RA, et al. Celecoxib reduces microvessel density in patients treated with nasopharyngeal carcinoma and induces changes in gene expression. Annals of Oncology. 2006;17(11):1625–1630. doi: 10.1093/annonc/mdl283. [DOI] [PubMed] [Google Scholar]
  • 64.Zhou Y, et al. Effect of celecoxib on E-cadherin, VEGF, Microvessel density and apoptosis in gastric cancer. Cancer Biology & Therapy. 2007;6(2):269–275. doi: 10.4161/cbt.6.2.3629. [DOI] [PubMed] [Google Scholar]
  • 65.Ruegg C, et al. The quest for surrogate markers of angiogenesis: A paradigm for translational research in tumor angiogenesis and anti-angiogenesis trials. Current Molecular Medicine. 2003;3(8):673–691. doi: 10.2174/1566524033479410. [DOI] [PubMed] [Google Scholar]
  • 66.Schor AM, et al. Heterogeneity in microvascular density in lung tumours: Comparison with normal bronchus. British Journal of Cancer. 1998;77(6):946–951. doi: 10.1038/bjc.1998.156. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Dales JP, et al. Prognostic significance of angiogenesis evaluated by CD105 expression compared to CD31 in 905 breast carcinomas: Correlation with long-term patient outcome. International Journal of Oncology. 2004;24(5):1197–1204. [PubMed] [Google Scholar]
  • 68.Duff SE, et al. CD105 is important for angiogenesis: Evidence and potential applications. The FASEB Journal. 2003;17(9):984–992. doi: 10.1096/fj.02-0634rev. [DOI] [PubMed] [Google Scholar]
  • 69.Kumar S, et al. Breast carcinoma: Vascular density determined using CD105 antibody correlates with tumor prognosis. Cancer Research. 1999;59(4):856–861. [PubMed] [Google Scholar]
  • 70.Tanaka F, et al. Evaluation of angiogenesis in non-small cell lung cancer: Comparison between anti-CD34 antibody and anti-CD105 antibody. Clinical Cancer Research. 2001;7(11):3410–3415. [PubMed] [Google Scholar]
  • 71.Sharma S, Sharma MC, Sarkar C. Morphology of angiogenesis in human cancer: A conceptual overview, histo-prognostic perspective and significance of neoangiogenesis. Histopathology. 2005;46(5):481–489. doi: 10.1111/j.1365-2559.2005.02142.x. [DOI] [PubMed] [Google Scholar]
  • 72.Algire GH, Chalkley HW, Legallais FY, Park HD. Vascular reactions of normal and malignant tissues in vivo. I. Vascular reactions of mice to wounds and to normal and neoplastic transplants. Journal of the National Cancer Institute. 1945;6:73–85. [Google Scholar]
  • 73.Vogel AW. Intratumoral vascular changes with increased size of a mammary adenocarcinoma: New method and results. Journal of the National Cancer Institute. 1965;34:571–578. [PubMed] [Google Scholar]
  • 74.Hilmas DE, Gillette EL. Tumor microvasculature following fractionated x irradiation. Radiology. 1975;116(1):165–169. doi: 10.1148/116.1.165. [DOI] [PubMed] [Google Scholar]
  • 75.Kadambi A, et al. Vascular endothelial growth factor (VEGF)-C differentially affects tumor vascular function and leukocyte recruitment: Role of VEGF-receptor 2 and host VEGF-A. Cancer Research. 2001;61(6):2404–2408. [PubMed] [Google Scholar]
  • 76.Cho WC, Cheng CH. Oncoproteomics: Current trends and future perspectives. Expert Review of Proteomics. 2007;4(3):401–410. doi: 10.1586/14789450.4.3.401. [DOI] [PubMed] [Google Scholar]
  • 77.Mittal V, Nolan DJ. Genomics and proteomics approaches in understanding tumor angiogenesis. Expert Review of Molecular Diagnostics. 2007;7(2):133–147. doi: 10.1586/14737159.7.2.133. [DOI] [PubMed] [Google Scholar]
  • 78.Petricoin EF, Liotta LA. Proteomic approaches in cancer risk and response assessment. Trends in Molecular Medicine. 2004;10(2):59–64. doi: 10.1016/j.molmed.2003.12.006. [DOI] [PubMed] [Google Scholar]
  • 79.Alexander H, et al. Proteomic analysis to identify breast cancer biomarkers in nipple aspirate fluid. Clinical Cancer Research. 2004;10(22):7500–7510. doi: 10.1158/1078-0432.CCR-04-1002. [DOI] [PubMed] [Google Scholar]
  • 80.Bhattacharyya S, et al. Diagnosis of pancreatic cancer using serum proteomic profiling. Neoplasia. 2004;6(5):674–686. doi: 10.1593/neo.04262. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Petricoin EF, et al. Use of proteomic patterns in serum to identify ovarian cancer. Lancet. 2002;359(9306):572–577. doi: 10.1016/S0140-6736(02)07746-2. [DOI] [PubMed] [Google Scholar]
  • 82.Petricoin EF, 3rd, et al. Serum proteomic patterns for detection of prostate cancer. Journal of the National Cancer Institute. 2002;94(20):1576–1578. doi: 10.1093/jnci/94.20.1576. [DOI] [PubMed] [Google Scholar]
  • 83.Conrads TP, et al. Proteomic patterns as a diagnostic tool for early-stage cancer: A review of its progress to a clinically relevant tool. Molecular Diagnosis. 2004;8(2):77–85. doi: 10.1007/BF03260049. [DOI] [PubMed] [Google Scholar]
  • 84.Kuerer HM, et al. Association between ductal fluid proteomic expression profiles and the presence of lymph node metastases in women with breast cancer. Surgery. 2004;136(5):1061–1069. doi: 10.1016/j.surg.2004.04.011. [DOI] [PubMed] [Google Scholar]
  • 85.Pusztai L, et al. Pharmacoproteomic analysis of prechemotherapy and postchemotherapy plasma samples from patients receiving neoadjuvant or adjuvant chemotherapy for breast carcinoma. Cancer. 2004;100(9):1814–1822. doi: 10.1002/cncr.20203. [DOI] [PubMed] [Google Scholar]
  • 86.Bouamrani A, et al. Direct-tissue SELDI-TOF mass spectrometry analysis: A new application for clinical proteomics. Clinical Chemistry. 2006;52(11):2103–2106. doi: 10.1373/clinchem.2006.070979. [DOI] [PubMed] [Google Scholar]
  • 87.Hwang SI, et al. Direct cancer tissue proteomics: a method to identify candidate cancer biomarkers from formalin-fixed paraffin-embedded archival tissues. Oncogene. 2007;26(1):65–76. doi: 10.1038/sj.onc.1209755. [DOI] [PubMed] [Google Scholar]
  • 88.Irish JM, Kotecha N, Nolan GP. Mapping normal and cancer cell signalling networks: Towards single-cell proteomics. Nature Reviews. Cancer. 2006;6(2):146–155. doi: 10.1038/nrc1804. [DOI] [PubMed] [Google Scholar]
  • 89.Tarnok A, Bocsi J, Brockhoff G. Cytomics—importance of multimodal analysis of cell function and proliferation in oncology. Cell Proliferation. 2006;39(6):495–505. doi: 10.1111/j.1365-2184.2006.00407.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Ornstein DK, Petricoin EF., 3rd Proteomics to diagnose human tumors and provide prognostic information. Oncology (Williston Park) 2004;18(4):521–529. discussion 529–32. [PubMed] [Google Scholar]
  • 91.Reid JD, Parker CE, Borchers CH. Protein arrays for biomarker discovery. Current Opinion in Molecular Therapeutics. 2007;9(3):216–221. [PubMed] [Google Scholar]
  • 92.Adachi J, et al. The human urinary proteome contains more than 1500 proteins, including a large proportion of membrane proteins. Genome Biology. 2006;7(9):R80. doi: 10.1186/gb-2006-7-9-r80. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Venable JD, et al. Relative quantification of stable isotope labeled peptides using a linear ion trap-Orbitrap hybrid mass spectrometer. Analytical Chemistry. 2007;79(8):3056–3064. doi: 10.1021/ac062054i. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Imami K, et al. Simple on-line sample preconcentration technique for peptides based on dynamic pH junction in capillary electrophoresis-mass spectrometry. Journal of Chromatography A. 2007;1148(2):250–255. doi: 10.1016/j.chroma.2007.03.014. [DOI] [PubMed] [Google Scholar]
  • 95.Bruneel A, et al. Proteomics of human umbilical vein endothelial cells applied to etoposide-induced apoptosis. Proteomics. 2005;5(15):3876–3884. doi: 10.1002/pmic.200401239. [DOI] [PubMed] [Google Scholar]
  • 96.Chen R, et al. Pancreatic cancer proteome: The proteins that underlie invasion, metastasis, and immunologic escape. Gastroenterology. 2005;129(4):1187–1197. doi: 10.1053/j.gastro.2005.08.001. [DOI] [PubMed] [Google Scholar]
  • 97.Shen F, et al. Functional proteometrics for cell migration. Cytometry A. 2006;69(7):563–572. doi: 10.1002/cyto.a.20283. [DOI] [PubMed] [Google Scholar]
  • 98.Shen J, et al. Identification and validation of differences in protein levels in normal, premalignant, and malignant lung cells and tissues using high-throughput western array and immunohistochemistry. Cancer Research. 2006;66(23):11194–11206. doi: 10.1158/0008-5472.CAN-04-1444. [DOI] [PubMed] [Google Scholar]
  • 99.Thompson LP, Dong Y. Chronic hypoxia decreases endothelial nitric oxide synthase protein expression in fetal guinea pig hearts. Journal of the Society for Gynecologic Investigation. 2005;12(6):388–395. doi: 10.1016/j.jsgi.2005.04.011. [DOI] [PubMed] [Google Scholar]
  • 100.Oh P, et al. Subtractive proteomic mapping of the endothelial surface in lung and solid tumours for tissue-specific therapy. Nature. 2004;429(6992):629–635. doi: 10.1038/nature02580. [DOI] [PubMed] [Google Scholar]
  • 101.Mustafa DAN, et al. Identification of glioma neovascularization-related proteins by using MALDI-FTMS and Nano-LC fractionation to microdissected tumor vessels. Molecular & Cellular Proteomics. 2007;6(7):1147–1157. doi: 10.1074/mcp.M600295-MCP200. [DOI] [PubMed] [Google Scholar]
  • 102.Christian S, et al. Nucleolin expressed at the cell surface is a marker of endothelial cells in angiogenic blood vessels. The Journal of Cell Biology. 2003;163(4):871–878. doi: 10.1083/jcb.200304132. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Hu J, et al. Gene expression signature for angiogenic and nonangiogenic non-small-cell lung cancer. Oncogene. 2005;24(7):1212–1219. doi: 10.1038/sj.onc.1208242. [DOI] [PubMed] [Google Scholar]
  • 104.Croix BS, et al. Genes expressed in human tumor endothelium. Science. 2000;289(5482):1197–1202. doi: 10.1126/science.289.5482.1197. [DOI] [PubMed] [Google Scholar]
  • 105.Seaman S, et al. Genes that distinguish physiological and pathological angiogenesis. Cancer Cell. 2007;11(6):539–554. doi: 10.1016/j.ccr.2007.04.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Li J-L, Harris AL. The potential of new tumor endothelium-specific markers for the development of antivascular therapy. Cancer Cell. 2007;11(6):478–481. doi: 10.1016/j.ccr.2007.05.004. [DOI] [PubMed] [Google Scholar]
  • 107.Nanda A, Croix B. Tumor endothelial markers: New targets for cancer therapy. Current Opinion in Oncology. 2004;16(1):44–49. doi: 10.1097/00001622-200401000-00009. [DOI] [PubMed] [Google Scholar]
  • 108.Beaty R, et al. PLXDC1 (TEM7) is identified in a genome-wide expression screen of glioblastoma endothelium. Journal of Neuro-Oncology. 2007;81(3):241–248. doi: 10.1007/s11060-006-9227-9. [DOI] [PubMed] [Google Scholar]
  • 109.Madden SL, et al. Vascular gene expression in nonneoplastic and malignant brain. The American Journal of Pathology. 2004;165(2):601–608. doi: 10.1016/s0002-9440(10)63324-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Parker BS, et al. Alterations in vascular gene expression in invasive breast carcinoma. Cancer Research. 2004;64(21):7857–7866. doi: 10.1158/0008-5472.CAN-04-1976. [DOI] [PubMed] [Google Scholar]
  • 111.Ho M, et al. Identification of endothelial cell genes by combined database mining and microarray analysis. Physiological Genomics. 2003;13(3):249–262. doi: 10.1152/physiolgenomics.00186.2002. [DOI] [PubMed] [Google Scholar]
  • 112.Yang X, Sun X. Meta-analysis of several gene lists for distinct types of cancer: A simple way to reveal common prognostic markers. BMC Bioinformatics. 2007;8(1):118. doi: 10.1186/1471-2105-8-118. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Costouros NG, et al. Microarray gene expression analysis of murine tumor heterogeneity defined by dynamic contrast-enhanced MRI. Molecular Imaging. 2002;1(3):301–308. doi: 10.1162/15353500200202124. [DOI] [PubMed] [Google Scholar]
  • 114.Jackson A, et al. Imaging tumor vascular heterogeneity and angiogenesis using dynamic contrast-enhanced magnetic resonance imaging. Clinical Cancer Research. 2007;13(12):3449–3459. doi: 10.1158/1078-0432.CCR-07-0238. [DOI] [PubMed] [Google Scholar]
  • 115.Menard C, et al. An interventional magnetic resonance imaging technique for the molecular characterization of intra-prostatic dynamic contrast enhancement. Molecular Imaging. 2005;4(1):63–66. doi: 10.1162/15353500200504181. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Jaeger J, et al. Gene expression signatures for tumor progression, tumor subtype, and tumor thickness in laser-microdissected melanoma tissues. Clinical Cancer Research. 2007;13(3):806–815. doi: 10.1158/1078-0432.CCR-06-1820. [DOI] [PubMed] [Google Scholar]
  • 117.Pen A, et al. Molecular markers of extracellular matrix remodeling in glioblastoma vessels: Microarray study of laser-captured glioblastoma vessels. Glia. 2007;55(6):559–572. doi: 10.1002/glia.20481. [DOI] [PubMed] [Google Scholar]
  • 118.Schuetz CS, et al. Progression-specific genes identified by expression profiling of matched ductal carcinomas in situ and invasive breast tumors, combining laser capture microdissection and oligonucleotide microarray analysis. Cancer Research. 2006;66(10):5278–5286. doi: 10.1158/0008-5472.CAN-05-4610. [DOI] [PubMed] [Google Scholar]
  • 119.Yang F, et al. Laser microdissection and microarray analysis of breast tumors reveal ER-alpha related genes and pathways. Oncogene. 2006;25(9):1413–1419. doi: 10.1038/sj.onc.1209165. [DOI] [PubMed] [Google Scholar]
  • 120.Scappaticci FA, et al. Surgical wound healing complications in metastatic colorectal cancer patients treated with bevacizumab. Journal of Surgical Oncology. 2005;91(3):173–180. doi: 10.1002/jso.20301. [DOI] [PubMed] [Google Scholar]
  • 121.Drevs J, Schneider V. The use of vascular biomarkers and imaging studies in the early clinical development of anti-tumour agents targeting angiogenesis. Journal of Internal Medicine. 2006;260(6):517–529. doi: 10.1111/j.1365-2796.2006.01727.x. [DOI] [PubMed] [Google Scholar]
  • 122.Zhang D, et al. Effects of a monoclonal anti-avb3 integrin antibody on blood vessels—A pharmacodynamic study. Investigational New Drugs. 2007;25(1):49–55. doi: 10.1007/s10637-006-9013-8. [DOI] [PubMed] [Google Scholar]
  • 123.Mundhenke C, et al. Tissue examination to monitor antiangiogenic therapy: A phase I clinical trial with endostatin. Clinical Cancer Research. 2001;7(11):3366–3374. [PubMed] [Google Scholar]
  • 124.Lockhart AC, et al. Reduction of wound angiogenesis in patients treated with BMS-275291, a broad spectrum matrix metalloproteinase inhibitor. Clinical Cancer Research. 2003;9(2):586–593. [PubMed] [Google Scholar]
  • 125.Secord A. Alvarez, et al. The relationship between serum vascular endothelial growth factor, persistent disease, and survival at second-look laparotomy in ovarian cancer. Gynecologic Oncology. 2004;94(1):74–79. doi: 10.1016/j.ygyno.2004.03.043. [DOI] [PubMed] [Google Scholar]
  • 126.Braybrooke JP, et al. A phase II study of razoxane, an antiangiogenic topoisomerase II inhibitor, in renal cell cancer with assessment of potential surrogate markers of angiogenesis. Clinical Cancer Research. 2000;6(12):4697–4704. [PubMed] [Google Scholar]
  • 127.Chan LW, et al. Urinary VEGF and MMP levels as predictive markers of 1-year progression-free survival in cancer patients treated with radiation therapy: A longitudinal study of protein kinetics throughout tumor progression and therapy. Journal of Clinical Oncology. 2004;22(3):499–506. doi: 10.1200/JCO.2004.07.022. [DOI] [PubMed] [Google Scholar]
  • 128.Coskun U, et al. Significance of serum vascular endothelial growth factor, insulin-like growth factor-I levels and nitric oxide activity in breast cancer patients. Breast. 2003;12(2):104–110. doi: 10.1016/s0960-9776(02)00214-x. [DOI] [PubMed] [Google Scholar]
  • 129.Duque JL, et al. Measurement of plasma levels of vascular endothelial growth factor in prostate cancer patients: Relationship with clinical stage, Gleason score, prostate volume, and serum prostate-specific antigen. Clinics. 2006;61(5):401–408. doi: 10.1590/s1807-59322006000500006. [DOI] [PubMed] [Google Scholar]
  • 130.Fine HA, et al. Phase II trial of the antiangiogenic agent thalidomide in patients with recurrent high-grade gliomas. Journal of Clinical Oncology. 2000;18(4):708–715. doi: 10.1200/JCO.2000.18.4.708. [DOI] [PubMed] [Google Scholar]
  • 131.Kaya A, et al. The prognostic significance of vascular endothelial growth factor levels in sera of non-small cell lung cancer patients. Respiratory Medicine. 2004;98(7):632–636. doi: 10.1016/j.rmed.2003.12.017. [DOI] [PubMed] [Google Scholar]
  • 132.Krzystek-Korpacka M, et al. Up-regulation of VEGF-C secreted by cancer cells and not VEGF-A correlates with clinical evaluation of lymph node metastasis in esophageal squamous cell carcinoma (ESCC) Cancer Letters. 2007;249(2):171–177. doi: 10.1016/j.canlet.2006.08.011. [DOI] [PubMed] [Google Scholar]
  • 133.Li L, et al. Correlation of serum VEGF levels with clinical stage, therapy efficacy, tumor metastasis and patient survival in ovarian cancer. Anticancer Research. 2004;24(3b):1973–1979. [PubMed] [Google Scholar]
  • 134.Poon RT, et al. Prognostic significance of serum vascular endothelial growth factor and endostatin in patients with hepatocellular carcinoma. The British Journal of Surgery. 2004;91(10):1354–1360. doi: 10.1002/bjs.4594. [DOI] [PubMed] [Google Scholar]
  • 135.Poon RT-P, et al. Quantitative correlation of serum levels and tumor expression of vascular endothelial growth factor in patients with hepatocellular carcinoma. Cancer Research. 2003;63(12):3121–3126. [PubMed] [Google Scholar]
  • 136.Shariat SF, et al. Association of preoperative plasma levels of vascular endothelial growth factor and soluble vascular cell adhesion molecule-1 with lymph node status and biochemical progression after radical prostatectomy. Journal of Clinical Oncology. 2004;22(9):1655–1663. doi: 10.1200/JCO.2004.09.142. [DOI] [PubMed] [Google Scholar]
  • 137.Shariat SF, et al. Association of pre- and postoperative plasma levels of transforming growth factor beta(1) and interleukin 6 and its soluble receptor with prostate cancer progression. Clinical Cancer Research. 2004;10(6):1992–1999. doi: 10.1158/1078-0432.ccr-0768-03. [DOI] [PubMed] [Google Scholar]
  • 138.Sliutz G, et al. Serum evaluation of basic FGF in breast cancer patients. Anticancer Research. 1995;15(6B):2675–2677. [PubMed] [Google Scholar]
  • 139.Tamura M, et al. Chest CT and serum vascular endothelial growth factor-C level to diagnose lymph node metastasis in patients with primary non-small cell lung cancer. Chest. 2004;126(2):342–346. doi: 10.1378/chest.126.2.342. [DOI] [PubMed] [Google Scholar]
  • 140.Gonzalez FJ, et al. Prognostic value of serum angiogenic activity in colorectal cancer patients. Journal of Cellular and Molecular Medicine. 2007;11(1):120–128. doi: 10.1111/j.1582-4934.2007.00005.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141.Negrier S, et al. Interleukin-6, interleukin-10, and vascular endothelial growth factor in metastatic renal cell carcinoma: Prognostic value of interleukin-6-from the Groupe Francais d’Immunotherapie. Journal of Clinical Oncology. 2004;22(12):2371–2378. doi: 10.1200/JCO.2004.06.121. [DOI] [PubMed] [Google Scholar]
  • 142.Tas F, et al. Serum vascular endothelial growth factor (VEGF) and bcl-2 levels in advanced stage non-small cell lung cancer. Cancer Investigation. 2006;24(6):576–580. doi: 10.1080/07357900600894781. [DOI] [PubMed] [Google Scholar]
  • 143.Brostjan C, et al. Monitoring of circulating angiogenic factors in dendritic cell-based cancer immunotherapy. Cancer. 2003;98(10):2291–2301. doi: 10.1002/cncr.11776. [DOI] [PubMed] [Google Scholar]
  • 144.Norden-Zfoni A, et al. Blood-based biomarkers of SU11248 activity and clinical outcome in patients with metastatic imatinib-resistant gastrointestinal stromal tumor. Clinical Cancer Research. 2007;13(9):2643–2650. doi: 10.1158/1078-0432.CCR-06-0919. [DOI] [PubMed] [Google Scholar]
  • 145.Drevs J, et al. Phase I clinical study of AZD2171, an oral vascular endothelial growth factor signaling inhibitor, in patients with advanced solid tumors. Journal of Clinical Oncology. 2007;25(21):3045–3054. doi: 10.1200/JCO.2006.07.2066. [DOI] [PubMed] [Google Scholar]
  • 146.Drevs J. Soluble markers for the detection of hypoxia under antiangiogenic treatment. Anticancer Research. 2003;23(2A):1159–1161. [PubMed] [Google Scholar]
  • 147.Vincenzi B, et al. Circulating VEGF reduction, response and outcome in advanced colorectal cancer patients treated with cetuximab plus irinotecan. Pharmacogenomics. 2007;8(4):319–327. doi: 10.2217/14622416.8.4.319. [DOI] [PubMed] [Google Scholar]
  • 148.Ria R, et al. Serum levels of angiogenic cytokines decrease after antineoplastic radiotherapy. Cancer Letters. 2004;216(1):103–107. doi: 10.1016/j.canlet.2004.05.022. [DOI] [PubMed] [Google Scholar]
  • 149.Shimada H, et al. Expression of angiogenic factors predicts response to chemoradiotherapy and prognosis of oesophageal squamous cell carcinoma. British Journal of Cancer. 2002;86(4):552–557. doi: 10.1038/sj.bjc.6600129. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 150.Shimada H, et al. Clinical significance of serum vascular endothelial growth factor in esophageal squamous cell carcinoma. Cancer. 2001;92(3):663–669. doi: 10.1002/1097-0142(20010801)92:3<663::aid-cncr1368>3.0.co;2-l. [DOI] [PubMed] [Google Scholar]
  • 151.Bewick M, et al. Evaluation of sICAM-1, sVCAM-1, and sE-selectin levels in patients with metastatic breast cancer receiving high-dose chemotherapy. Stem Cells and Development. 2004;13(3):281–294. doi: 10.1089/154732804323099217. [DOI] [PubMed] [Google Scholar]
  • 152.Ding YB, et al. Association of VCAM-1 over-expression with oncogenesis, tumor angiogenesis and metastasis of gastric carcinoma. World Journal of Gastroenterology. 2003;9(7):1409–1414. doi: 10.3748/wjg.v9.i7.1409. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 153.Kumar H, et al. Soluble FLT-1 is detectable in the sera of colorectal and breast cancer patients. Anticancer Research. 2002;22(3):1877–1880. [PubMed] [Google Scholar]
  • 154.Opala T, et al. Evaluation of soluble intracellular adhesion molecule-1 (sICAM-1) in benign and malignant ovarian masses. European Journal of Gynaecological Oncology. 2003;24(3–4):255–257. [PubMed] [Google Scholar]
  • 155.Pasieka Z, et al. Soluble intracellular adhesion molecules (sICAM-1, sVCAM-1) in peripheral blood of patients with thyroid cancer. Neoplasma. 2004;51(1):34–37. [PubMed] [Google Scholar]
  • 156.Pasieka Z, et al. Evaluation of the levels of bFGF, VEGF, sICAM-1, and sVCAM-1 in serum of patients with thyroid cancer. Recent Results in Cancer Research. 2003;162:189–194. doi: 10.1007/978-3-642-59349-9_18. [DOI] [PubMed] [Google Scholar]
  • 157.Ishii Y, Kitamura S. Soluble intercellular adhesion molecule-1 as an early detection marker for radiation pneumonitis. The European Respiratory Journal. 1999;13(4):733–738. doi: 10.1034/j.1399-3003.1999.13d06.x. [DOI] [PubMed] [Google Scholar]
  • 158.Nordal RA, Wong CS. Intercellular adhesion molecule-1 and blood-spinal cord barrier disruption in central nervous system radiation injury. Journal of Neuropathology and Experimental Neurology. 2004;63(5):474–483. doi: 10.1093/jnen/63.5.474. [DOI] [PubMed] [Google Scholar]
  • 159.Religa P, et al. Presence of bone marrow-derived circulating progenitor endothelial cells in the newly formed lymphatic vessels. Blood. 2005;106(13):4184–4190. doi: 10.1182/blood-2005-01-0226. [DOI] [PubMed] [Google Scholar]
  • 160.Jansen M, et al. Current perspectives on antiangiogenesis strategies in the treatment of malignant gliomas. Brain Research. Brain Research Reviews. 2004;45(3):143–163. doi: 10.1016/j.brainresrev.2004.03.001. [DOI] [PubMed] [Google Scholar]
  • 161.Lyden D, et al. Impaired recruitment of bone-marrow-derived endothelial and hematopoietic precursor cells blocks tumor angiogenesis and growth. Nature Medicine. 2001;7(11):1194–1201. doi: 10.1038/nm1101-1194. [DOI] [PubMed] [Google Scholar]
  • 162.Capillo M, et al. Continuous infusion of endostatin inhibits differentiation, mobilization, and clonogenic potential of endothelial cell progenitors. Clinical Cancer Research. 2003;9(1):377–382. [PubMed] [Google Scholar]
  • 163.Shaked Y, et al. Genetic heterogeneity of the vasculogenic phenotype parallels angiogenesis: Implications for cellular surrogate marker analysis of antiangiogenesis. Cancer Cell. 2005;7(1):101–111. doi: 10.1016/j.ccr.2004.11.023. [DOI] [PubMed] [Google Scholar]
  • 164.Gill M, et al. Vascular trauma induces rapid but transient mobilization of VEGFR2(+)AC133(+) endothelial precursor cells. Circulation Research. 2001;88(2):167–174. doi: 10.1161/01.res.88.2.167. [DOI] [PubMed] [Google Scholar]
  • 165.Shintani S, et al. Mobilization of endothelial progenitor cells in patients with acute myocardial infarction. Circulation. 2001;103(23):2776–2779. doi: 10.1161/hc2301.092122. [DOI] [PubMed] [Google Scholar]
  • 166.Rabascio C, et al. Assessing tumor angiogenesis: increased circulating VE-cadherin RNA in patients with cancer indicates viability of circulating endothelial cells. Cancer Research. 2004;64(12):4373–4377. doi: 10.1158/0008-5472.CAN-04-0265. [DOI] [PubMed] [Google Scholar]
  • 167.Furstenberger G, et al. Circulating endothelial cells and angiogenic serum factors during neoadjuvant chemotherapy of primary breast cancer. British Journal of Cancer. 2006;94(4):524–531. doi: 10.1038/sj.bjc.6602952. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 168.Ocak I, et al. The biologic basis of in vivo angiogenesis imaging. Frontiers in Bioscience. 2007;12:3601–3616. doi: 10.2741/2337. [DOI] [PubMed] [Google Scholar]
  • 169.Provenzale JM. Imaging of angiogenesis: Clinical techniques and novel imaging methods. AJR. American Journal of Roentgenology. 2007;188(1):11–23. doi: 10.2214/AJR.06.0280. [DOI] [PubMed] [Google Scholar]
  • 170.Schirner M, et al. Molecular imaging of tumor angiogenesis. Annals of the New York Academy of Sciences. 2004;1014(1):67–75. doi: 10.1196/annals.1294.007. [DOI] [PubMed] [Google Scholar]
  • 171.Herbst RS, et al. Development of biologic markers of response and assessment of antiangiogenic activity in a clinical trial of human recombinant endostatin. Journal of Clinical Oncology. 2002;20(18):3804–3814. doi: 10.1200/JCO.2002.05.102. [DOI] [PubMed] [Google Scholar]
  • 172.Jennens R, et al. Complete radiological and metabolic response of metastatic renal cell carcinoma to SU5416 (semaxanib) in a patient with probable von Hippel—Lindau syndrome. Urologic Oncology. 2004;22(3):193–196. doi: 10.1016/j.urolonc.2004.01.011. [DOI] [PubMed] [Google Scholar]
  • 173.Mullamitha SA, et al. Phase I evaluation of a fully human anti-{alpha}v integrin monoclonal antibody (CNTO 95) in patients with advanced solid tumors. Clinical Cancer Research. 2007;13(7):2128–2135. doi: 10.1158/1078-0432.CCR-06-2779. [DOI] [PubMed] [Google Scholar]
  • 174.Turner CD, et al. Phase II study of thalidomide and radiation in children with newly diagnosed brain stem gliomas and glioblastoma multiforme. Journal of Neurooncology. 2007;82(1):95–101. doi: 10.1007/s11060-006-9251-9. [DOI] [PubMed] [Google Scholar]
  • 175.Willett CG, et al. Complete pathological response to bevacizumab and chemoradiation in advanced rectal cancer. Nature Clinical Practice. Oncology. 2007;4(5):316–321. doi: 10.1038/ncponc0813. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 176.Clavo A, Brown R, Wahl R. Fluorodeoxyglucose uptake in human cancer cell lines is increased by hypoxia. Journal of Nuclear Medicine. 1995;36(9):1625–1632. [PubMed] [Google Scholar]
  • 177.Sivitz WI, et al. Pretranslational regulation of two cardiac glucose transporters in rats exposed to hypobaric hypoxia. American Journal of Physiology: Endocrinology and Metabolism. 1992;263(3):E562–E569. doi: 10.1152/ajpendo.1992.263.3.E562. [DOI] [PubMed] [Google Scholar]
  • 178.McDonald DM, Choyke PL. Imaging of angiogenesis: From microscope to clinic. Nature Medicine. 2003;9(6):713–725. doi: 10.1038/nm0603-713. [DOI] [PubMed] [Google Scholar]
  • 179.Eschmann SM, et al. Prognostic impact of hypoxia imaging with 18F-misonidazole PET in non-small cell lung cancer and head and neck cancer before radiotherapy. Journal of Nuclear Medicine. 2005;46(2):253–260. [PubMed] [Google Scholar]
  • 180.Gagel B, et al. [18F] fluoromisonidazole and [18F] fluorodeoxyglucose positron emission tomography in response evaluation after chemo-/radiotherapy of non-small-cell lung cancer: a feasibility study. BMC Cancer. 2006;6:51. doi: 10.1186/1471-2407-6-51. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 181.Laking GR, Price PM. Positron emission tomographic imaging of angiogenesis and vascular function. British Journal of Radiology. 2003;76(suppl1):S50–S59. doi: 10.1259/bjr/30399077. [DOI] [PubMed] [Google Scholar]
  • 182.Thorwarth D, et al. A kinetic model for dynamic [18F]-Fmiso PET data to analyse tumour hypoxia. Physics in Medicine & Biology. 2005;50(10):2209–2224. doi: 10.1088/0031-9155/50/10/002. [DOI] [PubMed] [Google Scholar]
  • 183.Thorwarth D, et al. Kinetic analysis of dynamic 18F-fluoromisonidazole PET correlates with radiation treatment outcome in head-and-neck cancer. BMC Cancer. 2005;5:152. doi: 10.1186/1471-2407-5-152. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 184.Cher LM, et al. Correlation of hypoxic cell fraction and angiogenesis with glucose metabolic rate in gliomas using 18F-fluoromisonidazole, 18F-FDG PET, and Immunohistochemical Studies. Journal of Nuclear Medicine. 2006;47(3):410–418. [PubMed] [Google Scholar]
  • 185.Hicks RJ, et al. Utility of FMISO PET in advanced head and neck cancer treated with chemoradiation incorporating a hypoxia-targeting chemotherapy agent. European Journal of Nuclear Medicine and Molecular Imaging. 2005;32(12):1384–1391. doi: 10.1007/s00259-005-1880-2. [DOI] [PubMed] [Google Scholar]
  • 186.Rischin D, et al. Prognostic significance of [18F]-misonidazole positron emission tomography-detected tumor hypoxia in patients with advanced head and neck cancer randomly assigned to chemoradiation with or without tirapazamine: A substudy of Trans-Tasman Radiation Oncology Group Study 98.02. Journal of Clinical Oncology. 2006;24(13):2098–2104. doi: 10.1200/JCO.2005.05.2878. [DOI] [PubMed] [Google Scholar]
  • 187.Rischin D, et al. Phase I trial of concurrent tirapazamine, cisplatin, and radiotherapy in patients with advanced head and neck cancer. Journal of Clinical Oncology. 2001;19(2):535–542. doi: 10.1200/JCO.2001.19.2.535. [DOI] [PubMed] [Google Scholar]
  • 188.Pal A, et al. Molecular imaging of EGFR kinase activity in tumors with 124I-labeled small molecular tracer and positron emission tomography. Molecular Imaging and Biology. 2006;8(5):262–277. doi: 10.1007/s11307-006-0049-0. [DOI] [PubMed] [Google Scholar]
  • 189.Schmidt K, et al. Angiostatin overexpression in Morris hepatoma results in decreased tumor growth but increased perfusion and vascularization. Journal of Nuclear Medicine. 2006;47(3):543–551. [PubMed] [Google Scholar]
  • 190.Anderson H, et al. Measurement of renal tumour and normal tissue perfusion using positron emission tomography in a phase II clinical trial of razoxane. British Journal of Cancer. 2003;89(2):262–267. doi: 10.1038/sj.bjc.6601105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 191.Lehtio K, et al. Imaging perfusion and hypoxia with PET to predict radiotherapy response in head-and-neck cancer. International Journal of Radiation Oncology, Biology, Physics. 2004;59(4):971–982. doi: 10.1016/j.ijrobp.2003.12.014. [DOI] [PubMed] [Google Scholar]
  • 192.Mullani N, et al. First pass FDG measured blood flow in tumors: A comparison with O-15 labeled water measured blood flow. Clinical Positron Imaging. 2000;3(4):153. doi: 10.1016/s1095-0397(00)00065-0. [DOI] [PubMed] [Google Scholar]
  • 193.Tseng J, et al. 18F-FDG kinetics in locally advanced breast cancer: Correlation with tumor blood flow and changes in response to neoadjuvant chemotherapy. Journal of Nuclear Medicine. 2004;45(11):1829–1837. [PubMed] [Google Scholar]
  • 194.Wells P, et al. Assessment of proliferation in vivo using 2-[11C]thymidine positron emission tomography in advanced intra-abdominal malignancies. Cancer Research. 2002;62(20):5698–5702. [PubMed] [Google Scholar]
  • 195.Chen X, et al. Pegylated Arg-Gly-Asp peptide: 64Cu labeling and PET imaging of brain tumor {alpha}v{beta}3-integrin expression. Journal of Nuclear Medicine. 2004;45(10):1776–1783. [PubMed] [Google Scholar]
  • 196.Chen X, et al. MicroPET imaging of breast cancer alphav-integrin expression with 64Cu-labeled dimeric RGD peptides. Molecular Imaging and Biology. 2004;6(5):350–359. doi: 10.1016/j.mibio.2004.06.004. [DOI] [PubMed] [Google Scholar]
  • 197.Chen X, et al. MicroPET imaging of brain tumor angiogenesis with 18F-labeled PEGylated RGD peptide. European Journal of Nuclear Medicine and Molecular Imaging. 2004;31(8):1081–1089. doi: 10.1007/s00259-003-1452-2. [DOI] [PubMed] [Google Scholar]
  • 198.Collingridge DR, et al. The Development of [124I] Iodinated-VG76e: A novel tracer for imaging vascular endothelial growth factor in vivo using positron emission tomography. Cancer Research. 2002;62(20):5912–5919. [PubMed] [Google Scholar]
  • 199.Furumoto S, et al. Tumor detection using 18F-labeled matrix metalloproteinase-2 inhibitor. Nuclear Medicine and Biology. 2003;30(2):119–125. doi: 10.1016/s0969-8051(02)00393-1. [DOI] [PubMed] [Google Scholar]
  • 200.Haubner R, et al. Glycosylated RGD-containing peptides: Tracer for tumor targeting and angiogenesis imaging with improved biokinetics. Journal of Nuclear Medicine. 2001;42(2):326–336. [PubMed] [Google Scholar]
  • 201.Kobayashi H, et al. Application of a macromolecular contrast agent for detection of alterations of tumor vessel permeability induced by radiation. Clinical Cancer Research. 2004;10(22):7712–7720. doi: 10.1158/1078-0432.CCR-04-1175. [DOI] [PubMed] [Google Scholar]
  • 202.Zheng Q, et al. Synthesis, biodistribution and micro-PET imaging of a potential cancer biomarker carbon-11 labeled MMP inhibitor (2R)-2-[[4-(6-fluorohex-1-ynyl)phenyl]sulfonylamino]-3-methylbutyric acid [11C]methyl ester. Nuclear Medicine and Biology. 2003;30(7):753–760. doi: 10.1016/s0969-8051(03)00086-6. [DOI] [PubMed] [Google Scholar]
  • 203.Zinn K, et al. Imaging Tc-99m-labeled FGF-1 targeting in rats. Nuclear Medicine and Biology. 2000;27(4):407–414. doi: 10.1016/s0969-8051(00)00090-1. [DOI] [PubMed] [Google Scholar]
  • 204.Cai W, et al. PET of vascular endothelial growth factor receptor expression. Journal of Nuclear Medicine. 2006;47(12):2048–2056. [PubMed] [Google Scholar]
  • 205.Wang H, et al. A new PET tracer specific for vascular endothelial growth factor receptor 2. European Journal of Nuclear Medicine and Molecular Imaging. 2007;34(12):2001–2010. doi: 10.1007/s00259-007-0524-0. [DOI] [PubMed] [Google Scholar]
  • 206.Beer AJ, et al. Biodistribution and pharmacokinetics of the {alpha}v{beta}3-selective tracer 18F-Galacto-RGD in cancer patients. Journal of Nuclear Medicine. 2005;46(8):1333–1341. [PubMed] [Google Scholar]
  • 207.Wu Z, et al. MicroPET of tumor integrin {alpha}v{beta}3 expression using 18F-labeled PEGylated tetrameric RGD peptide (18F-FPRGD4) Journal of Nuclear Medicine. 2007;48(9):1536–1544. doi: 10.2967/jnumed.107.040816. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 208.Kamel E, et al. CT vs 68Ge attenuation correction in a combined PET/CT system: evaluation of the effect of lowering the CT tube current. European Journal of Nuclear Medicine and Molecular Imaging. 2002;29(3):346–350. doi: 10.1007/s00259-001-0698-9. [DOI] [PubMed] [Google Scholar]
  • 209.Antoch G, et al. Accuracy of whole-body dual-modality fluorine-18–2-Fluoro-2-Deoxy-D-Glucose Positron Emission Tomography and Computed Tomography (FDG-PET/CT) for tumor staging in solid tumors: Comparison with CT and PET. Journal of Clinical Oncology. 2004;22(21):4357–4368. doi: 10.1200/JCO.2004.08.120. [DOI] [PubMed] [Google Scholar]
  • 210.Gorres G, Steinert H, Schulthess G. v. PET and functional anatomic fusion imaging in lung and breast cancers. The Cancer Journal. 2004;10(4):251–261. doi: 10.1097/00130404-200407000-00006. [DOI] [PubMed] [Google Scholar]
  • 211.Barrett T, et al. MRI of tumor angiogenesis. Journal of Magnetic Resonance Imaging. 2007;26(2):235–249. doi: 10.1002/jmri.20991. [DOI] [PubMed] [Google Scholar]
  • 212.Hylton N. Dynamic contrast-enhanced magnetic resonance imaging as an imaging biomarker. Journal of Clinical Oncology. 2006;24(20):3293–3298. doi: 10.1200/JCO.2006.06.8080. [DOI] [PubMed] [Google Scholar]
  • 213.Choyke P, Dwyer A, Knopp M. Functional tumor imaging with dynamic contrast-enhanced magnetic resonance imaging. Journal of Magnetic Resonance Imaging. 2003;17(5):509–520. doi: 10.1002/jmri.10304. [DOI] [PubMed] [Google Scholar]
  • 214.Padhani A, Dzik-Jurasz A. Perfusion MR imaging of extracranial tumor angiogenesis. Topics in Magnetic Resonance Imaging. 2004;15(1):41–57. doi: 10.1097/00002142-200402000-00005. [DOI] [PubMed] [Google Scholar]
  • 215.Kiessling F, Morgenstern B, Zhang C. Contrast agents and applications to assess tumor angiogenesis in vivo by magnetic resonance imaging. Current Medicinal Chemistry. 2007;14(1):77–91. doi: 10.2174/092986707779313516. [DOI] [PubMed] [Google Scholar]
  • 216.Daldrup HE, et al. Quantification of the extraction fraction for gadopentetate across breast cancer capillaries. Magnetic Resonance in Medicine. 1998;40(4):537–543. doi: 10.1002/mrm.1910400406. [DOI] [PubMed] [Google Scholar]
  • 217.Padhani A. Dynamic contrast-enhanced MRI in clinical oncology: Current status and future directions. Journal of Magnetic Resonance Imaging. 2002;16(4):407–422. doi: 10.1002/jmri.10176. [DOI] [PubMed] [Google Scholar]
  • 218.Wikstrom M, et al. Contrast-enhanced MRI of tumors. Comparison of Gd-DTPA and a macromolecular agent. Investigative Radiology. 1989;24(8):609–615. doi: 10.1097/00004424-198908000-00007. [DOI] [PubMed] [Google Scholar]
  • 219.Kang H, et al. Magnetic resonance imaging of inducible E-selectin expression in human endothelial cell culture. Bio-conjugate Chemistry. 2002;13(1):122–127. doi: 10.1021/bc0155521. [DOI] [PubMed] [Google Scholar]
  • 220.McCarthy JR, et al. Targeted delivery of multifunctional magnetic nanoparticles. Nanomed. 2007;2(2):153–167. doi: 10.2217/17435889.2.2.153. [DOI] [PubMed] [Google Scholar]
  • 221.Sancey L, et al. In vivo imaging of tumour angiogenesis in mice with the alpha(v)beta (3) integrin-targeted tracer (99m) Tc-RAFT-RGD. European Journal of Nuclear Medicine and Molecular Imaging. 2007;34:2037–2047. doi: 10.1007/s00259-007-0497-z. [DOI] [PubMed] [Google Scholar]
  • 222.Sipkins D, et al. Detection of tumor angiogenesis in vivo by alphaVbeta3-targeted magnetic resonance imaging. Nature Medicine. 1998;4(5):623–626. doi: 10.1038/nm0598-623. [DOI] [PubMed] [Google Scholar]
  • 223.Winter PM, et al. Molecular imaging of angiogenesis in nascent Vx-2 rabbit tumors using a novel {alpha}{nu}{beta} 3-targeted nanoparticle and 1.5 tesla magnetic resonance imaging. Cancer Research. 2003;63(18):5838–5843. [PubMed] [Google Scholar]
  • 224.Nunn A, Linder K, Tweedle M. Can receptors be imaged with MRI agents? The Quarterly Journal of Nuclear Medicine. 1997;41(2):155–162. [PubMed] [Google Scholar]
  • 225.O’Donnell A, et al. A Phase I study of the angiogenesis inhibitor SU5416 (semaxanib) in solid tumours, incorporating dynamic contrast MR pharmacodynamic end points. British Journal of Cancer. 2005;93(8):876–883. doi: 10.1038/sj.bjc.6602797. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 226.Kothari M, et al. Imaging in antiangiogenesis trial: a clinical trials radiology perspective. British Journal of Radiology. 2003;76(suppl1):S92–96. doi: 10.1259/bjr/25400972. [DOI] [PubMed] [Google Scholar]
  • 227.Jain RK. Normalizing tumor vasculature with anti-angiogenic therapy: A new paradigm for combination therapy. Nature Medicine. 2001;7(9):987–989. doi: 10.1038/nm0901-987. [DOI] [PubMed] [Google Scholar]
  • 228.Tatum JL, Hoffman JM. Role of imaging in clinical trials of antiangiogenesis therapy in oncology. Academic Radiology. 2000;7(10):798–799. doi: 10.1016/s1076-6332(00)80628-5. [DOI] [PubMed] [Google Scholar]
  • 229.Medved M, et al. Semiquantitative analysis of dynamic contrast enhanced MRI in cancer patients: Variability and changes in tumor tissue over time. Journal of Magnetic Resonance Imaging. 2004;20(1):122–128. doi: 10.1002/jmri.20061. [DOI] [PubMed] [Google Scholar]
  • 230.Morgan B, et al. Dynamic contrast-enhanced magnetic resonance imaging as a biomarker for the pharmacological response of PTK787/ZK 222584, an inhibitor of the vascular endothelial growth factor receptor tyrosine kinases, in patients with advanced colorectal cancer and liver metastases: Results from two Phase I studies. Journal of Clinical Oncology. 2003;21(21):3955–3964. doi: 10.1200/JCO.2003.08.092. [DOI] [PubMed] [Google Scholar]
  • 231.Xiong HQ, et al. A phase I surrogate endpoint study of SU6668 in patients with solid tumors. Investigational New Drugs. 2004;22(4):459–466. doi: 10.1023/B:DRUG.0000036688.96453.8d. [DOI] [PubMed] [Google Scholar]
  • 232.Robinson SP, et al. Susceptibility contrast magnetic resonance imaging determination of fractional tumor blood volume: A noninvasive imaging biomarker of response to the vascular disrupting agent ZD6126. International Journal of Radiation Oncology, Biology, Physics. 2007;69(3):872–879. doi: 10.1016/j.ijrobp.2007.06.061. [DOI] [PubMed] [Google Scholar]
  • 233.Liu G, et al. Dynamic contrast-enhanced magnetic resonance imaging as a pharmacodynamic measure of response after acute dosing of AG-013736, an oral angiogenesis inhibitor, in patients with advanced solid tumors: Results from a Phase I study. Journal of Clinical Oncology. 2005;23(24):5464–5473. doi: 10.1200/JCO.2005.04.143. [DOI] [PubMed] [Google Scholar]
  • 234.Akella NS, et al. Assessment of brain tumor angiogenesis inhibitors using perfusion magnetic resonance imaging: Quality and analysis results of a phase I trial. Journal of Magnetic Resonance Imaging. 2004;20(6):913–922. doi: 10.1002/jmri.20202. [DOI] [PubMed] [Google Scholar]
  • 235.Nabors LB, et al. Phase I and correlative biology study of cilengitide in patients with recurrent malignant glioma. Journal of Clinical Oncology. 2007;25(13):1651–1657. doi: 10.1200/JCO.2006.06.6514. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 236.Thomas AL, et al. Phase I study of the safety, tolerability, pharmacokinetics, and pharmacodynamics of PTK787/ZK 222584 administered twice daily in patients with advanced cancer. Journal of Clinical Oncology. 2005;23(18):4162–4171. doi: 10.1200/JCO.2005.09.034. [DOI] [PubMed] [Google Scholar]
  • 237.Cuenod CA, et al. Tumor angiogenesis: Pathophysiology and implications for contrast-enhanced MRI and CT assessment. Abdominal Imaging. 2006;31(2):188–193. doi: 10.1007/s00261-005-0386-5. [DOI] [PubMed] [Google Scholar]
  • 238.Lee TY, Purdie TG, Stewart E. CT imaging of angiogenesis. The Quarterly Journal of Nuclear Medicine. 2003;47(3):171–187. [PubMed] [Google Scholar]
  • 239.Miles KA. Functional CT imaging in oncology. European Radiology. 2003;13(Suppl 5):M134–M138. doi: 10.1007/s00330-003-2108-0. [DOI] [PubMed] [Google Scholar]
  • 240.Tateishi U, et al. Contrast-enhanced dynamic computed tomography for the evaluation of tumor angiogenesis in patients with lung carcinoma. Cancer. 2002;95(4):835–842. doi: 10.1002/cncr.10730. [DOI] [PubMed] [Google Scholar]
  • 241.Tateishi U, et al. Tumor angiogenesis and dynamic CT in lung adenocarcinoma: radiologic—pathologic correlation. Journal of Computer Assisted Tomography. 2001;25(1):23–27. doi: 10.1097/00004728-200101000-00004. [DOI] [PubMed] [Google Scholar]
  • 242.Yi CA, et al. Solitary pulmonary nodules: Dynamic enhanced multi-detector row CT study and comparison with vascular endothelial growth factor and microvessel density. Radiology. 2004;233(1):191–199. doi: 10.1148/radiol.2331031535. [DOI] [PubMed] [Google Scholar]
  • 243.Ma S-H, et al. Peripheral lung cancer: Relationship between multi-slice spiral CT perfusion imaging and tumor angiogenesis and cyclin D1 expression. Clinical Imaging. 2007;31(3):165–177. doi: 10.1016/j.clinimag.2007.01.011. [DOI] [PubMed] [Google Scholar]
  • 244.Faria SC, et al. CT quantification of effects of thalidomide in patients with metastatic renal cell carcinoma. American Journal of Roentgenology. 2007;189(2):378–385. doi: 10.2214/AJR.07.2164. [DOI] [PubMed] [Google Scholar]
  • 245.McNeel DG, et al. Phase I trial of a monoclonal antibody specific for alphavbeta3 integrin (MEDI-522) in patients with advanced malignancies, including an assessment of effect on tumor perfusion. Clinical Cancer Research. 2005;11(21):7851–7860. doi: 10.1158/1078-0432.CCR-05-0262. [DOI] [PubMed] [Google Scholar]
  • 246.Ng Q-S, et al. Effect of nitric-oxide synthesis on tumour blood volume and vascular activity: A phase I study. The Lancet Oncology. 2007;8(2):111–118. doi: 10.1016/S1470-2045(07)70001-3. [DOI] [PubMed] [Google Scholar]
  • 247.Xiong HQ, et al. A phase I surrogate endpoint study of SU6668 in patients with solid tumors. Investigational New Drugs. 2004;22(4):459–466. doi: 10.1023/B:DRUG.0000036688.96453.8d. [DOI] [PubMed] [Google Scholar]
  • 248.Kim J, et al. Solitary pulmonary nodules: A comparative study evaluated with contrast-enhanced dynamic MR imaging and CT. Journal of Computer Assisted Tomography. 2004;28(6):766–775. doi: 10.1097/00004728-200411000-00007. [DOI] [PubMed] [Google Scholar]
  • 249.Korst R, Altorki N. Imaging for esophageal tumors. Thoracic Surgery Clinics. 2004;14(1):61–69. doi: 10.1016/S1547-4127(04)00038-6. [DOI] [PubMed] [Google Scholar]
  • 250.Kramer H, et al. Oesophageal endoscopic ultrasound with fine needle aspiration improves and simplifies the staging of lung cancer. Thorax. 2004;59(7):596–601. doi: 10.1136/thx.2003.018028. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 251.Massari M, et al. Value and limits of endorectal ultrasonography for preoperative staging of rectal carcinoma. Surgical Laparoscopy & Endoscopy. 1998;8(6):438–444. [PubMed] [Google Scholar]
  • 252.Saga Y, et al. Comparative study of novel endoluminal ultrasonography and conventional transurethral ultrasonography in staging of bladder cancer. International Journal of Urology. 2004;11(8):597–601. doi: 10.1111/j.1442-2042.2004.00865.x. [DOI] [PubMed] [Google Scholar]
  • 253.Tarantino D, Bernstein M. Endoanal ultrasound in the staging and management of squamous-cell carcinoma of the anal canal: Potential implications of a new ultrasound staging system. Diseases of the Colon & Rectum. 2002;45(1):16–22. doi: 10.1007/s10350-004-6108-1. [DOI] [PubMed] [Google Scholar]
  • 254.Yanai H, et al. Prognostic value and interobserver agreement of endoscopic ultrasonography for superficial squamous cell carcinoma of the esophagus: A prospective study. International Journal of Gastrointestinal Cancer. 2003;34(1):1–8. doi: 10.1385/IJGC:34:1:01. [DOI] [PubMed] [Google Scholar]
  • 255.Cosgrove D. Angiogenesis imaging—ultrasound. British Journal of Radiology. 2003;76(suppl1):S43–S49. doi: 10.1259/bjr/86364648. [DOI] [PubMed] [Google Scholar]
  • 256.Huber S, et al. Breast tumors: computer-assisted quantitative assessment with color Doppler US. Radiology. 1994;192(3):797–801. doi: 10.1148/radiology.192.3.8058950. [DOI] [PubMed] [Google Scholar]
  • 257.Fanelli M, et al. Assessment of tumor vascularization: Immunohistochemical and non-invasive methods. The International Journal of Biological Markers. 1999;14(4):218–231. doi: 10.1177/172460089901400405. [DOI] [PubMed] [Google Scholar]
  • 258.Dayton P, et al. Ultrasonic analysis of peptide- and antibody-targeted microbubble contrast agents for molecular imaging of alphavbeta3-expressing cells. Molecular Imaging and Biology. 2004;3(2):125–134. doi: 10.1162/1535350041464883. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 259.Harrer J, et al. Perfusion imaging of high-grade gliomas: A comparison between contrast harmonic and magnetic resonance imaging. Technical note. Journal of Neurosurgery. 2004;101(4):700–703. doi: 10.3171/jns.2004.101.4.0700. [DOI] [PubMed] [Google Scholar]
  • 260.Kiessling F, et al. Comparing dynamic parameters of tumor vascularization in nude mice revealed by magnetic resonance imaging and contrast-enhanced intermittent power Doppler sonography. Investigative Radiology. 2003;38(8):516–524. doi: 10.1097/01.rli.0000073448.16334.fe. [DOI] [PubMed] [Google Scholar]
  • 261.Abdollahi A, et al. Combined therapy with direct and indirect angiogenesis inhibition results in enhanced antiangiogenic and antitumor effects. Cancer Research. 2003;63(24):8890–8898. [PubMed] [Google Scholar]
  • 262.Forsberg F, et al. Assessment of angiogenesis: Implications for ultrasound imaging. Ultrasonics. 2004;42(1–9):325–330. doi: 10.1016/j.ultras.2003.12.026. [DOI] [PubMed] [Google Scholar]
  • 263.Fury M, et al. A Phase II study of SU5416 in patients with advanced or recurrent head and neck cancers. Investigational New Drugs. 2007;25(2):165–172. doi: 10.1007/s10637-006-9011-x. [DOI] [PubMed] [Google Scholar]
  • 264.Mross K, et al. Phase I clinical and pharmacokinetic study of PTK/ZK, a multiple VEGF receptor inhibitor, in patients with liver metastases from solid tumours. European Journal of Cancer. 2005;41(9):1291–1299. doi: 10.1016/j.ejca.2005.03.005. [DOI] [PubMed] [Google Scholar]
  • 265.Mross K, Fuxius S, Drevs J. Serial measurements of pharmacokinetics, DCE-MRI, blood flow, PET and bio-markers in serum/plasma—what is a useful tool in clinical studies of anti-angiogenic drugs? International Journal of Clinical Pharmacology and Therapeutics. 2002;40(12):573–574. doi: 10.5414/cpp40573. [DOI] [PubMed] [Google Scholar]
  • 266.Gibson AP, Hebden JC, Arridge SR. Recent advances in diffuse optical imaging. Physics in Medicine & Biology. 2005;50(4):R1–R43. doi: 10.1088/0031-9155/50/4/r01. [DOI] [PubMed] [Google Scholar]
  • 267.Cai W, Chen X. Multimodality imaging of vascular endothelial growth factor and vascular endothelial growth factor receptor expression. Frontiers in Bioscience. 2007;12:4267–4279. doi: 10.2741/2386. [DOI] [PubMed] [Google Scholar]
  • 268.Chance B, et al. Breast cancer detection based on incremental biochemical and physiological properties of breast cancers: A six-year, two-site Study1. Academic Radiology. 2005;12(8):925–933. doi: 10.1016/j.acra.2005.04.016. [DOI] [PubMed] [Google Scholar]
  • 269.Zhu Q, et al. Benign versus malignant breast masses: Optical differentiation with US-guided optical imaging reconstruction. Radiology. 2005;237(1):57–66. doi: 10.1148/radiol.2371041236. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 270.Ntziachristos V, Chance B. Probing physiology and molecular function using optical imaging: applications to breast cancer. Breast Cancer Research. 2001;3(1):41–46. doi: 10.1186/bcr269. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 271.Escudier B, et al. Sorafenib in advanced clear-cell renal-cell carcinoma. The New England Journal of Medicine. 2007;356(2):125–134. doi: 10.1056/NEJMoa060655. [DOI] [PubMed] [Google Scholar]
  • 272.Giantonio BJ, et al. Bevacizumab in combination with oxaliplatin, fluorouracil, and leucovorin (FOLFOX4) for previously treated metastatic colorectal cancer: Results from the Eastern Cooperative Oncology Group Study E3200. Journal of Clinical Oncology. 2007;25(12):1539–1544. doi: 10.1200/JCO.2006.09.6305. [DOI] [PubMed] [Google Scholar]
  • 273.Hurwitz H, et al. Bevacizumab plus irinotecan, fluorouracil, and leucovorin for metastatic colorectal cancer. The New England Journal of Medicine. 2004;350(23):2335–2342. doi: 10.1056/NEJMoa032691. [DOI] [PubMed] [Google Scholar]
  • 274.Sandler A, et al. Paclitaxel—carboplatin alone or with bevacizumab for non-small-cell lung cancer. The New England Journal of Medicine. 2006;355(24):2542–2550. doi: 10.1056/NEJMoa061884. [DOI] [PubMed] [Google Scholar]
  • 275.Bonner JA, et al. Radiotherapy plus cetuximab for squamous-cell carcinoma of the head and neck. The New England Journal of Medicine. 2006;354(6):567–578. doi: 10.1056/NEJMoa053422. [DOI] [PubMed] [Google Scholar]
  • 276.Cunningham D, et al. Cetuximab monotherapy and cetuximab plus irinotecan in irinotecan-refractory metastatic colorectal cancer. The New England Journal of Medicine. 2004;351(4):337–345. doi: 10.1056/NEJMoa033025. [DOI] [PubMed] [Google Scholar]
  • 277.Gibson TB, Ranganathan A, Grothey A. Randomized phase III trial results of panitumumab, a fully human anti-epidermal growth factor receptor monoclonal antibody, in metastatic colorectal cancer. Clinical Colorectal Cancer. 2006;6(1):29–31. doi: 10.3816/CCC.2006.n.01. [DOI] [PubMed] [Google Scholar]
  • 278.Van Cutsem E, et al. Open-label Phase III trial of panitumumab plus best supportive care compared with best supportive care alone in patients with chemotherapy-refractory metastatic colorectal cancer. Journal of Clinical Oncology. 2007;25(13):1658–1664. doi: 10.1200/JCO.2006.08.1620. [DOI] [PubMed] [Google Scholar]
  • 279.Piccart-Gebhart MJ, et al. Trastuzumab after adjuvant chemotherapy in HER2-positive breast cancer. The New England Journal of Medicine. 2005;353(16):1659–1672. doi: 10.1056/NEJMoa052306. [DOI] [PubMed] [Google Scholar]
  • 280.Moore MJ, et al. Erlotinib plus gemcitabine compared with gemcitabine alone in patients with advanced pancreatic cancer: A Phase III trial of the National Cancer Institute of Canada Clinical Trials Group. Journal of Clinical Oncology. 2007;25(15):1960–1966. doi: 10.1200/JCO.2006.07.9525. [DOI] [PubMed] [Google Scholar]
  • 281.Shepherd FA, et al. Erlotinib in previously treated non-small-cell lung cancer. The New England Journal of Medicine. 2005;353(2):123–132. doi: 10.1056/NEJMoa050753. [DOI] [PubMed] [Google Scholar]
  • 282.Thatcher N, et al. Gefitinib plus best supportive care in previously treated patients with refractory advanced non-small-cell lung cancer: Results from a randomised, placebo-controlled, multicentre study (Iressa Survival Evaluation in Lung Cancer) Lancet. 2005;366(9496):1527–1537. doi: 10.1016/S0140-6736(05)67625-8. [DOI] [PubMed] [Google Scholar]
  • 283.Dagher R, et al. Approval summary: Imatinib mesylate in the treatment of metastatic and/or unresectable malignant gastrointestinal stromal tumors. Clinical Cancer Research. 2002;8(10):3034–3038. [PubMed] [Google Scholar]
  • 284.Druker BJ, et al. Five-year follow-up of patients receiving imatinib for chronic myeloid leukemia. The New England Journal of Medicine. 2006;355(23):2408–2417. doi: 10.1056/NEJMoa062867. [DOI] [PubMed] [Google Scholar]
  • 285.Geyer CE, et al. Lapatinib plus capecitabine for HER2-positive advanced breast cancer. The New England Journal of Medicine. 2006;355(26):2733–2743. doi: 10.1056/NEJMoa064320. [DOI] [PubMed] [Google Scholar]
  • 286.Casali PG, et al. Updated results from a phase III trial of sunitinib in GIST patients (pts) for whom imatinib (IM) therapy has failed due to resistance or intolerance. Journal of Clinical Oncology (Meeting Abstracts) 2006;24(18suppl):9513. [Google Scholar]
  • 287.Motzer RJ, et al. Sunitinib versus interferon alfa in metastatic renal-cell carcinoma. The New England Journal of Medicine. 2007;356(2):115–124. doi: 10.1056/NEJMoa065044. [DOI] [PubMed] [Google Scholar]
  • 288.Orlowski RZ, et al. Randomized phase III study of pegylated liposomal doxorubicin plus bortezomib compared with bortezomib alone in relapsed or refractory multiple myeloma: Combination therapy improves time to progression. Journal of Clinical Oncology. 2007;25(25):3892–901. doi: 10.1200/JCO.2006.10.5460. [DOI] [PubMed] [Google Scholar]
  • 289.Richardson PG, et al. Bortezomib or high-dose dexamethasone for relapsed multiple myeloma. The New England Journal of Medicine. 2005;352(24):2487–2498. doi: 10.1056/NEJMoa043445. [DOI] [PubMed] [Google Scholar]
  • 290.Guilhot F, et al. Interferon alfa-2b combined with cytarabine versus interferon alone in chronic myelogenous leukemia. French Chronic Myeloid Leukemia Study Group. The New England Journal of Medicine. 1997;337(4):223–229. doi: 10.1056/NEJM199707243370402. [DOI] [PubMed] [Google Scholar]
  • 291.Hauschild A, et al. Prospective randomized trial of interferon Alfa-2b and Interleukin-2 as adjuvant treatment for resected intermediate- and high-risk primary melanoma without clinically detectable node metastasis. Journal of Clinical Oncology. 2003;21(15):2883–2888. doi: 10.1200/JCO.2003.07.116. [DOI] [PubMed] [Google Scholar]
  • 292.Hudes G, et al. Temsirolimus, interferon alfa, or both for advanced renal-cell carcinoma. The New England Journal of Medicine. 2007;356(22):2271–2281. doi: 10.1056/NEJMoa066838. [DOI] [PubMed] [Google Scholar]
  • 293.Barlogie B, et al. Thalidomide and hematopoietic-cell transplantation for multiple myeloma. The New England Journal of Medicine. 2006;354(10):1021–1030. doi: 10.1056/NEJMoa053583. [DOI] [PubMed] [Google Scholar]
  • 294.Weber DM, et al. Lenalidomide plus high-dose dexamethasone provides improved overall survival compared to high-dose dexamethasone alone for relapsed or refractory multiple myeloma (MM): Results of a North American phase III study (MM-009) Journal of Clinical Oncology (Meeting Abstracts) 2006;24(18suppl):7521. [Google Scholar]

RESOURCES