REVIEW

Clinical Application of Antiangiogenic Therapy: Microvessel Density, What It Does and Doesn't Tell Us

Lynn Hlatky, Philip Hahnfeldt, Judah Folkman

Affiliations of authors: L. Hlatky, P. Hahnfeldt, Dana-Farber Cancer Institute and the Department of Radiation Oncology, Harvard Medical School, Boston, MA; J. Folkman, Laboratory of Surgical Research, Department of Surgery, Children's Hospital, Boston, and Departments of Cell Biology and Surgery, Harvard Medical School.

Correspondence to: Lynn Hlatky, Ph.D., JFB-523, Dana-Farber Cancer Institute, Harvard Medical School, 44 Binney St., Boston, MA 02115 (e-mail: Lynn_Hlatky{at}dfci.harvard.edu).


    INTRODUCTION
 Top
 Notes
 Introduction
 Microvessel Density Is A...
 Microvessel Density May Not...
 Looking Beyond Measurements of...
 References
 
A substantial number of clinical trials using antiangiogenic therapies are ongoing worldwide. How to achieve the maximum benefit from these therapies and how to monitor patient response are of paramount concern to investigators. There are currently no markers of the net angiogenic activity of a tumor available to aid investigators in the design of antiangiogenic treatment schemes. It stands to reason that quantification of various aspects of tumor vasculature might provide an indication of angiogenic activity. One often-quantified aspect of tumor vasculature is microvessel density. Studies over the last decade have demonstrated the value of using tumor microvessel density as a prognostic indicator for a wide range of cancers. In this context, measurement of microvessel density facilitates assessments of disease stage and the likelihood of recurrence and helps guide treatment decisions. Recently, however, it has been assumed by some investigators that measurements of microvessel density may also reveal the degree of angiogenic activity in a tumor. Based on this assumption, quantification of microvessel density is thought to constitute a surrogate marker for the efficacy of antiangiogenic agents as well as a means by which to assess which patients are good candidates for antiangiogenic therapy prior to treatment. Here we contend that, although microvessel density is a useful prognostic marker, it is not, by itself, an indicator of therapeutic efficacy, nor should it be used to guide the stratification of patients for therapeutic trials. In this review, we discuss the evidence for these assertions and what can and cannot be determined from measurements of microvessel density.


    MICROVESSEL DENSITY IS A USEFUL PROGNOSTIC INDICATOR
 Top
 Notes
 Introduction
 Microvessel Density Is A...
 Microvessel Density May Not...
 Looking Beyond Measurements of...
 References
 
As early as 1972, Brem and colleagues (1) saw the need to define a quantitative measure of tumor angiogenesis and proposed a grading system for human brain tumors that relied on the analysis of endothelial cell characteristics in conjunction with vessel density measurements. With the advent of specific antibodies to detect endothelial cells, quantitative studies of tumor vascularization intensified. In the early 1990s, Weidner et al. (24) showed that measurement of microvessel density within isolated regions of high vessel concentration (i.e., hotspots) was a prognostic indicator for human breast and prostate carcinomas. In this capacity, microvessel density measurements aid in assessing the stage of disease; the likelihood of metastasis, recurrence, or survival; and the planning of treatment course (5,6). Since those early studies, hundreds of reports have examined the prognostic value of microvessel density in breast, prostate, and other cancers. Most of these studies report positive correlations between microvessel density and tumor recurrence, although some report no or even negative correlations between these endpoints. Some of the discrepancies may be explained by the fact that details of the methodology used to assay microvessel density can influence its value as a prognostic indicator. For example, the choice of the antibody (e.g., CD34, CD31, von Willebrand factor, or CD105) used to detect endothelial cells of the tumor microvasculature (79) has been reported to influence the study outcome, as does whether microvessel density is assessed at the periphery or center of the tumor (10). Despite these technical issues, microvessel density as measured by the hotspot method of Weidner (24) is a valuable prognostic indicator for a wide range of tumor types. The accumulated data indicating the prognostic value of microvessel density in breast cancer was recently reviewed by Gasparini (11). Earlier, Gasparini and Harris (12) reviewed published reports through 1999 on the prognostic value of microvessel density for a variety of solid tumors. We have summarized their compiled results and have reviewed the current literature for malignant melanoma (1319), non-small-cell lung (7,8,2034), genitourinary (4,9,3551), esophageal (5254), and gastrointestinal cancers (5565). These data are presented in Tables 1–4GoGoGoGo. Evidence is accumulating that measurements of microvessel density may also have predictive value for hematologic cancers (6671). Compiled in Table 5Go are the results of studies to date that have examined the prognostic value of microvessel density measurements in the bone marrow of patients with hematologic malignancies (67,68,71).


View this table:
[in this window]
[in a new window]
 
Table 1. Prognostic value of microvessel density measurements in malignant melanoma
 

View this table:
[in this window]
[in a new window]
 
Table 2. Prognostic value of microvessel density measurements in non-small-cell lung cancers
 

View this table:
[in this window]
[in a new window]
 
Table 3. Prognostic value of microvessel density measurements in genitourinary cancers
 

View this table:
[in this window]
[in a new window]
 
Table 4. Prognostic value of microvessel density measurements in esophageal and gastrointestinal tumors
 

View this table:
[in this window]
[in a new window]
 
Table 5. Prognostic value of microvessel density measurements in bone marrow for hematologic malignancies
 

    MICROVESSEL DENSITY MAY NOT BE AN INDICATOR OF ANTIANGIOGENIC TREATMENT EFFICACY
 Top
 Notes
 Introduction
 Microvessel Density Is A...
 Microvessel Density May Not...
 Looking Beyond Measurements of...
 References
 
Despite its importance as a prognostic indicator in untreated tumors, microvessel density has not been shown to be a valid measure to guide or evaluate antiangiogenic treatment. The presumptions that the degree of tumor microvessel density is equivalent to the degree of tumor angiogenic activity, and that the quantification of microvessel density will provide the much needed dictates for antiangiogenic therapy, have led to two serious misconceptions: 1) that one can use the level of microvessel density of the untreated tumor to decide whether a patient will respond to antiangiogenic therapy and, thus, which patients to include in clinical trials; and 2) that the efficacy of the antiangiogenic agent will necessarily be reflected by a decreasing microvessel density during the course of antiangiogenic treatment. Here we discuss these and other apparent misconceptions concerning microvessel density in the context of antiangiogenic therapy. The first five misconceptions concern the interpretation of microvessel density in untreated tumors, and the last two concern treated tumors.

1) Microvessel Density Is Not a Measure of the Angiogenic Dependence of a Tumor. To a Large Degree, It Reflects the Metabolic Burden of the Supported Tumor Cells.

Microvessel density varies widely with tumor type. The degree of variability in microvessel density among tumor types is often misconstrued to mean that some tumors depend on angiogenesis whereas others do not. All tumor types, even those with low microvessel densities, depend on a therapeutically targetable angiogenic process. This angiogenic dependence is based on the inviolable demand of a growing tumor for sufficient levels of nutrients and oxygen exchange (72). These metabolic requirements are in addition to any dependence of the tumor cells on paracrine factors provided by the endothelial cells (73,74). Even leukemia, considered to be a `liquid tumor,' has now been shown to induce angiogenesis in the bone marrow to support its growth (75). Evidence of the importance of angiogenesis in hematologic malignancies, in general, is rapidly accruing. Both myeloid and lymphoid disorders may be accompanied by an increase in the microvessel density of the bone marrow (6671,75).

Contrary to common belief, microvessel density does not reflect the angiogenic activity or angiogenic dependence of a tumor. Microvessel density is a measure of the number of vessels per high-power (microscope) field and, as such, reflects intercapillary distance. Intercapillary distances are determined at the local level by the net balance between angiogenic factors that stimulate and those that inhibit vessel growth in each micro-region, as well as by nonangiogenic factors, such as the oxygen and nutrient consumption rates of the tumor cells. Oxygen and nutrient consumption limit how far away from the vasculature tumor cells can remain viable and, thus, the number of tumor cells that can squeeze between capillaries before some become necrotic (Fig. 1Go). The metabolic needs of cancer cells vary with the tissue of origin and change with tumor progression. Thus, the number of tumor cells that can be supported by a vessel varies, influencing, in turn, the vascular density of the tumor. As was pointed out by Thomlinson and Gray (76), one can think of the supported tumor cells as forming a viable cuff around a vessel, with cuff size being roughly indicative of the metabolic burden of the cancer cells. This is well illustrated in the Dunning rat model of prostate carcinoma, shown in Fig. 2Go, wherein tumor cells within approximately 110 µm of the vasculature form a viable cuff around a functional tumor vessel. Outside this radius of oxygen and nutrient support, tumor cells cannot survive and an abrupt shift to necrosis is observed.



View larger version (38K):
[in this window]
[in a new window]
 
Fig. 1. Microvessel density reflects intercapillary distance. As this schematic of a tumor section with two vessels illustrates, intercapillary distance (indicated by the yellow arrow), and thus microvessel density, is influenced by both angiogenic and nonangiogenic factors. Microvessel density is therefore not a pure measure of angiogenic activity. FGF = fibroblast growth factor; VEGF = vascular endothelial growth factor; TGF-{beta} = transforming growth factor-{beta}; O2 = oxygen.

 


View larger version (167K):
[in this window]
[in a new window]
 
Fig. 2. Supported tumor cells forming cuffs (areas of viable tumor cells surrounding functional vessels—indicated by black dashed ovals) are shown for a Dunning rat prostate carcinoma xenograft. Cuff size is roughly indicative of the metabolic burden of the carcinoma cells. Tumor cells within approximately 110 µm of the vasculature are viable; beyond this radius of oxygen and nutrient support, an abrupt shift to necrosis is observed. Section was stained with hematoxylin for DNA, highlighting areas of necrosis, and with an antibody to CD31, showing the endothelium.

 
Cuff size tends to vary inversely with tumor metabolic demand. Tumors that have high rates of oxygen or nutrient consumption, such as glioblastomas, have small cuffs only two to three cells wide and have a high vascular density. By contrast, tumors of low metabolic demand, such as chondrosarcomas, have relatively large cuff sizes, with many cell layers supported and a relatively low vascular density. In normal tissues, cuff size also varies with metabolic demand. For example, in the liver, a tissue that has a high oxygen consumption rate, cuff size is one cell layer thick, and every hepatocyte is adjacent to an endothelial cell. In human tumors, the oxygen consumption rate is often substantially lower than that of the corresponding normal tissue (77). It is therefore consistent, albeit counterintuitive, that certain human tumor types, despite their often considerable angiogenic activity, can exhibit lower microvessel densities than the corresponding normal tissues. For example, the microvessel densities for human lung, mammary, renal cell, and colon carcinomas are lower than those of their normal tissue counterparts (78). In addition, the microvessel density for glioblastoma is less than that for brain tissue (78), and pituitary adenomas are less vascular than the normal pituitary gland (79). [Prostate tumors and germ cell tumors of the testis are two notable exceptions to this trend—these tumors show higher microvessel densities than their normal tissue counterparts (36,78,80)]. Thus, the fact that increases in microvessel density do not necessarily coincide with the onset of angiogenesis, a hallmark of neoplastic transformation (81), underscores the fact that microvessel density is neither a measure of tumor angiogenic activity nor a measure of tumor angiogenic dependence.

2) The Measurement of Microvessel Density Is Not Predictive of Tumor Response Under Antiangiogenic Treatment and Therefore Is Not Useful for Stratifying Patients for Clinical Trials. Low Microvessel Density Does Not Portend a Poor Response to Antiangiogenic Therapy.

In recent years there has been a trend toward individualizing tumor treatment by using predictive assays to select patients who are the most likely to respond to specific cancer therapies (8284). Predictive assays are used to evaluate to what degree an individual patient's tumor exhibits the specific attributes [e.g., the level of tumor hypoxia (84)] that a specialized cancer therapy (e.g., neutron radiotherapy) targets. It is a common misconception that antiangiogenic treatment can be applied only to cancers that have high microvessel densities. This thinking promotes the further misconceptions that the measurement of microvessel density is the relevant predictive assay for antiangiogenic therapy and that the level of microvascular density within untreated tumors forecasts the efficacy of tumor response to antiangiogenic treatment. Thus, it is widely assumed that tumors with high microvessel densities are good candidates for clinical trials of antiangiogenic therapies, whereas tumors that typically have low microvessel densities (e.g., astrocytomas) are thought to be poor candidates for such clinical trials (85). However, experimental evidence does not support this method of patient stratification, but rather shows that both poorly vascularized and highly vascularized tumors can respond to antiangiogenic therapy. For example, bladder tumors that had less vascularity were found to be treatable by lower doses of an antiangiogenic agent than were those that had more vascularity (86). Because virtually all tumors are angiogenesis-dependent (72,87), low microvessel counts within tumors are not sufficient criteria to exclude patients from treatment with angiogenesis inhibitors.

3) Tumor Microvessel Density May Not Vary in Accordance With the Tissue or Blood Levels of any Single Proangiogenic Factor.

Many studies have investigated the relationship between microvessel density and the levels of single angiogenic factors such as vascular endothelial growth factor (VEGF) (27,32). It is now clear that individual tumors can make a wide variety of proangiogenic factors, and the relative expressions of these factors can change over time. For example, when breast cancers were screened for their expression of a panel of angiogenic factors, the majority originally expressed only a single positive factor, either VEGF or acidic fibroblast growth factor (aFGF), but evolved over time to express up to six angiogenic stimulators (88). The net angiogenic influence of the tumor microenvironment should be thought of as the sum of the positive and negative regulators of angiogenesis that arise from both the tumor cells (89) and host tissues (90). Human breast cancer fibroblasts, for example, exhibit high levels of VEGF (90). Because it is the net effect of total angiogenic stimulation and inhibition that determines the level of neovascularization, the levels of single angiogenic factors are not, as a general rule, strictly associated with measures of tumor vascularization. This principle has been borne out by investigations showing no correlation between tumor microvessel density measurements and levels of VEGF and aFGF or basic fibroblast growth factor (bFGF) (9194). Such findings need to be considered when proposing antiangiogenic cancer therapies that seek to block a single proangiogenic signal. Recent work from the Kerbel group (95) showed that, when the proangiogenic factor VEGF is targeted by antiangiogenic therapy, tumors that express p53 respond more rapidly than tumors that do not. They interpreted the reduced response in the absence of p53 as resistance. More likely, p53-null status does not confer resistance. Rather, p53-null status is proangiogenic (96) through increased expressions of VEGF, interleukin 8 (IL-8), and bFGF, the suppression of which requires a suitably inclusive antiangiogenic counterattack.

4) A Minimum Vessel Density Is Determined by Tumor Cell Metabolic Demand, but Vessel Density Can Exceed the Metabolic Requirements of a Tumor.

Although normally functioning tissues rarely overvascularize, tumors can engage in angiogenic activity beyond that dictated by their metabolic needs, thus leading to overvascularization. As previously discussed, a major factor contributing to vessel density is metabolic demand. Metabolic demand places a lower limit on the density of vessels within the tissue required to maintain tissue viability. If vascular density should fall below the required minimum, the flux of oxygen and glucose to the cells will be inadequate, triggering just enough tumor cell death to restore the minimal vascular density. Attesting to this process, regions of necrotic tumor cells adjacent to, and resulting from, oxygen or nutrient shortages are seen in many human tumors (97) (see Fig. 2Go). In normal tissues, by contrast, the level of microvascular density fairly accurately reflects the metabolic demands of the cells. This is because evolutionary pressures have forced a tight and efficient coupling between vascular supply and metabolic need. In the genetically unstable tumor, the close coupling between vascular density and oxygen or nutrient consumption may be loosened. In tumors, angiogenic factor expression often becomes uncoupled from normal regulatory controls and, consequently, some angiogenic factors are constitutively expressed at high levels. A prime example is the dissociation of VEGF expression from its regulation by oxygen concentration; in normal tissues, VEGF expression is increased and VEGF mRNA is stabilized under conditions of hypoxia or ischemia (90,98,99), whereas in tumor cells, VEGF is often constitutively expressed at high levels regardless of the ambient oxygen tension (37,100,101). Notable examples of the decoupling of oxygen tension from VEGF expression occur in p53-null tumors (102) and in renal cell carcinomas from individuals that carry a mutation in the von Hippel-Lindau (VHL) gene (103). Both p53 and VHL genes are associated with the degradation of hypoxia-inducible factor-1{alpha}, an upstream regulator of VEGF.

5) Rapid Tumor Growth Does Not Imply High Vascular Density. The Microvessel Density of a Tumor Need Not Be Higher, and Is Often Lower, Than That of its Corresponding Normal Tissue, Which Is Experiencing no Net Growth.

Whereas microvessel density frequently increases during tumor progression to accommodate an increased metabolic demand or a decoupling of net angiogenic stimulator expression with metabolic need, microvessel density levels do not reflect growth rate. In fact, Eberhard et al. (78) have shown that for five of the six human cancers examined (prostate carcinoma being the exception), the microvessel density of the tumor was lower than that of the corresponding nongrowing normal tissue. In lung carcinoma, for example, microvessel density was found to be only 29% that of normal lung tissue. In glioblastoma, an exceptionally highly vascularized tumor, microvessel density was found to be 78% that of normal brain tissue. This apparent paradox can be partially explained by the lower oxygen consumption rate of tumor cells (77). In addition, tumor cells are known to tolerate oxygen deprivation and to be resistant to apoptosis under hypoxic conditions (104). Because tumor cells can remain viable at lower oxygen concentrations, they can exist at greater distances from the vasculature than can their normal-cell counterparts. Both the lowered oxygen consumption of tumor cells and their tolerance of hypoxic conditions promote increased intercapillary distance in tumors relative to their normal tissue counterparts.

Although the amount of total tumor vascularization (an extrinsic variable applicable to the tumor as a whole) must increase rapidly in fast-growing tumors to support a rapidly increasing tumor mass, the density of the vessels (an intrinsic variable that is locally defined) need not be high. In addition, the efficacy of antiangiogenic therapy would not be expected a priori to vary with the growth rate of the tumor. The lack of growth rate dependence for antiangiogenic effect stands in marked contrast to what is seen under standard chemotherapy. Chemotherapy targets proliferating cells directly and thus exerts a more demonstrable effect for fast-growing tumors than it does for slow-growing tumors.

6) The Efficacy of Antiangiogenic Agents Cannot Be Simply Visualized by Alterations in Microvessel Density During Treatment.

There is an urgent need to assess the clinical activity of the numerous antiangiogenic agents that are now in patient trials (105,106). Because antiangiogenic therapy suppresses tumor cell growth indirectly by inhibiting endothelial cell growth, reductions in tumor sizes or growth rates under antiangiogenic therapy are likely to occur over longer time frames than they would when standard chemotherapy is used (89). It follows that rapid tumor shrinkage, which is the classic end point scored for in clinical trials of chemotherapeutic agents, cannot be used to reliably discriminate between a null and a positive response to antiangiogenic agents. Instead, an appropriate measure of vascular inhibition is needed to assess the efficacy of an antiangiogenic agent at an early stage in a trial (i.e., before tumor growth inhibition is observed).

A common, albeit erroneous, impression is that measurements of microvessel density made during antiangiogenic therapy may be used to evaluate the therapeutic response to antiangiogenic agents. Numerous clinical protocols for antiangiogenic therapy include the periodic taking of biopsies to evaluate tumor microvessel density (107). However, the efficacy of an antiangiogenic agent cannot be evaluated by measuring the changes in microvessel density that the agent induces. This is because changes in microvessel density do not independently measure vascular inhibition, but rather, reflect the changing ratio of the vascular component of the tumor to its tumor-cell component. Under antiangiogenic therapy, capillary inhibition or elimination occurs first, followed by tumor-cell elimination, and both influence microvessel density. The tightness of the coupling of these two tumor components determines how much the vascular density fluctuates. Consequently, microvessel density is a time-dependent measure that varies in a nonmonotonic manner during antiangiogenic treatment—first decreasing in response to capillary inhibition or elimination, then decreasing more slowly or even increasing somewhat as tumor cells drop out. Finally, in those cases where the angiogenic inhibitor is cleared from the bloodstream following bolus dosing (89), vessel density may asymptotically approach the pretreatment microvessel density level if treatment does not alter the metabolic demand or the angiogenic factor production of the tumor cells. The time required to partially or fully restore microvessel density to its pretreatment level following the clearance of an administered bolus dose of an angiogenic inhibitor would depend on the tightness of the coupling between vessel dropout and tumor-cell dropout. This would vary for different tumor types. If the restoration time is less than the typical times between biopsies, one might expect that most measures of microvessel density that are made over the course of treatment would yield a value little changed from the pretreatment value.

Although decreases in vascular density certainly provide a rough indication of the activity of an antiangiogenic agent, the nonmonotonic nature of microvessel density under antiangiogenic therapy renders microvessel density unsuitable as a stand-alone measure to monitor antiangiogenic effect. The concept that measures of vascular density alone do not reflect the efficacy of angiogenic inhibitors is demonstrated in Fig. 3Go. An untreated tumor (control) and two tumors treated with the angiogenic inhibitor endostatin are shown. Despite the fact that the inhibitor substantially inhibited growth in both of the treated tumors, the post-treatment levels of vascularization of the two tumors vary substantially. Compared with the control tumor, vessel density was substantially lower in one treated tumor and slightly higher in the second. Thus, detection of a decrease in microvessel density during treatment with an antiangiogenic agent suggests that the agent is active. However, the absence of a drop in microvessel density does not indicate that the agent is ineffective.



View larger version (53K):
[in this window]
[in a new window]
 
Fig. 3. Microvessel density may not coincide with antiangiogenic treatment response. Human liposarcoma xenografts were untreated (Control) or treated with the angiogenic inhibitor endostatin (Endostatin Treated). Tumor masses were measured with calipers using the formula length x width x width/2, and differences of mean masses (n = 6) were quantified using Student's t test. Shown are representative tumors (photo insets), vascularization within those tumors as detected by an antibody to CD34 (a transmembrane glycoprotein constitutively expressed on endothelial cells), and vascular density as quantified by digitized imaging. Compared with the control tumor, the endostatin-treated tumors displayed statistically significantly (P<.001) less tumor growth, yielding a treated tumor to control tumor mass ratio (T/C) of 0.085. The post-treatment levels of vascularization of the two tumors shown varied considerably. Entire tumor sections were scored for vessel density by densitometric detection of CD34, which was assessed by imaging microscope fields at x200 across the entire section (ignoring necrotic regions). Microvessel density dropped sharply in one treated tumor (center histogram) but increased slightly in the second treated tumor (right histogram). The histograms show the proportion of microscope fields over the tumor sections that had the indicated ratios of vessel area/tumor area. A drop in microvessel density is reflected by a leftward shift of the histogram distribution relative to the control, because more fields show low ratios of vascularization relative to tumor area. A leftward shift of the distribution, and thus a drop in microvessel density, is seen for the endostatin-treated tumor in the middle but not for the endostatin-treated tumor on the right.

 
The lack of a parallel relationship between tumor size and tumor vessel density points to the error in interpreting the lack of a decrease in microvessel density during therapy as a failure to inhibit vascularization. Measuring slight decreases, no change, or even increases in microvessel density is still consistent with vessel inhibition because microvessel density is a dynamically complex quantity that is influenced by the initial vascular suppression plus the consequent interaction between the vascular and tumor-cell compartments (Fig. 4Go). A seminal clinical study illustrates this point. The study showed that multiple myeloma that was resistant to high-dose chemotherapy regressed in response to treatment with a single antiangiogenic agent, thalidomide (107), even though not all tumor regressions were accompanied by a decrease in microvessel density. This finding has prompted some skepticism about whether thalidomide acts via antiangiogenic mechanisms to promote tumor regression (107,108). However, the observation of tumor regression without a corresponding decrease in microvessel density does not indicate that mechanisms other than antiangiogenesis play a causal role in treatment response. The tumor cell population may simply decrease in direct proportion to, and as a direct consequence of, the loss of its supporting vasculature. The tendency to read too much into the behavior of microvessel density over the course of treatment of multiple myeloma patients with thalidomide further highlights the need for the development of appropriate surrogate markers to accurately assess angiogenic suppression in clinical trials. In a recent study, Bertolini et al. (109) reported that decreases in microvessel density in multiple myeloma patients who responded to thalidomide did not correlate with treatment response, although other measures of decreased angiogenic activity did correlate with treatment response in these same patients. Specifically, the levels of activated circulating endothelial cells derived from bone marrow decreased 10-fold after thalidomide treatment.



View larger version (36K):
[in this window]
[in a new window]
 
Fig. 4. Microvessel density in a shrinking tumor is determined by the combined effects of capillary dropout and tumor cell dropout. The time-dependent coupling of these two forms of cell dropout is reflected by the intercapillary distance and, in turn, by microvessel density. Thus, following antiangiogenic therapy a regressing tumor may exhibit decreased microvessel density, essentially unchanged levels of microvessel density, or even increased microvessel density.

 
7) All Tumor Microvessels Are Not Functionally Equal. Inhibition of Ineffective Vessels Has Little Consequence in the Reduction of Tumor Growth, Further Emphasizing the Lack of a Simple Quantitative Relationship Between Alterations in Microvessel Density and Tumor Inhibition During Antiangiogenic Treatment.

All tumor vessels are not equal in their ability to provide oxygen and nutrients to the tumor cells they support. Vessels may be inefficient for several reasons. Tumor vessels can themselves be hypoxic and carry little oxygen, or they can have oscillating rather than directed blood flows and thus be ineffective at transporting oxygen and nutrients. Clearly, inhibiting hypoxic vasculature would shrink a tumor mass less than would inhibiting oxygen-rich vessels. In addition, the vascularization of a tumor may be greater than necessary to support its metabolic needs, thereby creating a state of overvascularization. Inhibiting redundant vasculature would likewise be expected to shrink a tumor mass less than would inhibiting the more effective vessels. In these cases, however, there is benefit to extending treatment. For example, in an effectively overvascularized p53-null tumor, there may be an initial period during antiangiogenic therapy when there is little tumor suppression because the excess vasculature is being targeted. As therapy continues and more critical vasculature becomes the target, a shift to more marked suppression would be observed. This appears to be what is happening in the study by Yu et al. (95), which looked at the response of p53-null colon carcinoma to antiangiogenic therapy using vinblastine and DC101, an antibody specific to VEGF receptor-2 expressed on endothelium.

The disconnect between vascular reduction and tumor response points to the lack of a simple quantitative relationship between therapeutic knockout of capillaries, reduction of microvessel density, and measurable tumor regression. In current theoretical treatments of therapeutic response to antiangiogenic agents, the inability to attribute tumor response to local vessel densities because of inhomogeneities in transport capacities is being dealt with by considering only the "effective vasculature" feeding the tumor. By focusing specifically on the functional vessels in a tumor, the underlying dynamics connecting vessel inhibition and tumor regression are likely to be clarified (89).


    LOOKING BEYOND MEASUREMENTS OF MICROVESSEL DENSITY
 Top
 Notes
 Introduction
 Microvessel Density Is A...
 Microvessel Density May Not...
 Looking Beyond Measurements of...
 References
 
Switching the target of cancer treatment from the exceptionally heterogeneous tumor cell population (110) to the considerably more homogeneous tumor vasculature is a revolutionary therapeutic approach (87). In principle, this approach offers the means to treat cancers systemically by reaching both the primary tumor and its metastases (111) while circumventing, to large extent, three classic problems associated with chemotherapy: drug resistance (87,112,113), normal tissue toxicity (112), and limited access of drugs to target cells. A current challenge is to find appropriate characteristics of the vasculature that could serve as clinical markers to guide antiangiogenic treatment and dosing. In this regard, there has recently been heightened interest in the role that microvessel density may play in indicating therapeutic responsiveness. Quantifying microvessel density has proven to be a valuable independent prognostic indicator in a wide variety of human cancers (12) and, as such, has become a useful tool for assessing treatment options. Arguably, the prognostic value of microvessel density points to the importance of the mechanisms that drive increased vascularization (such as a shift in the balance of angiogenic factors in favor of stimulation) in the biology of tumor progression. On the other hand, measures of microvessel density are not sufficient to reveal the functional or angiogenic status of tumor neovasculature. Microvessel density, accordingly, offers no indication as to which patients might best respond to antiangiogenic therapy. In addition, although decreases in microvessel density following antiangiogenic treatment can give an indication of the antivascular activity of a particular agent, microvessel density as a single end point fails to provide an adequate measure for resolving the vascular response to antiangiogenic agents.

Although microvessel density per se would not influence the responsiveness of a tumor to antiangiogenic treatment, the detailed composition of the tumor vessels [e.g., the fraction of proliferating endothelial cells they contain (78) or whether pericytes are present (114)] might well modify the action of a particular antiangiogenic agent. VEGF neutralization (e.g., with VEGF-specific antibodies) may not affect mature vessels but has been shown to lead to the regression of immature vessels that lack smooth muscle cells (115). Assays that quantify the maturation state of vessels might also provide some indication of the susceptibility of the existing vasculature in the tumor bed to specific antiangiogenic agents. These assays include those that measure the ratio of concentrations of the angiopoietins Ang-2 and Ang-1 (116), or the presence or absence of an epitope recognized by the monoclonal antibody LH39 that is expressed in the lamina lucida and is associated with vessel maturity (117). Currently, there is a flurry of investigation into the fundamental biology of angiogenesis (118121) that will inevitably yield further insights into the effective use of antiangiogenic agents (122). With antiangiogenic therapy, the challenge comes not in restricting the patient pool, but in providing the proper guidelines for using combination therapies [e.g., those that use several antiangiogenic agents in combination or combined with radiation (123,124) or chemotherapy (125,126)]; delivering the agents [e.g., extended or continuous versus bolus dosing schemes (89,125129)]; classifying the different types of antiangiogenic agents according to target (115), mode of action, or stage of cancer most amenable to antiangiogenic agent attack (130); unveiling and exploiting the serendipitous antiangiogenic effects of classic chemotherapeutic agents (125,126,131136); and accurately assessing patient response, including the identification of appropriate surrogate end points. Recognizing the limitations of microvessel density as a surrogate end point will no doubt improve our resolve to explore supplementary assays for evaluating the efficacy of antiangiogenic agents.


    NOTES
 Top
 Notes
 Introduction
 Microvessel Density Is A...
 Microvessel Density May Not...
 Looking Beyond Measurements of...
 References
 
Supported by Public Health Service (PHS) grants CA86302 (to L. Hlatky), CA78496 (to P. Hahnfeldt), and CA64481 (to J. Folkman) from the National Cancer Institute, National Institutes of Health, Department of Health and Human Services (DHHS); a grant from the Gerry and Nancy Pencer Brain Trust (to L. Hlatky); and a grant from EntreMed, Inc. (Rockville, MD) (to Children's Hospital).

We thank Dipak Panigrahy and Samuel Singer for human liposarcoma xenografts, Lloyd Hutchinson for Dunning rat prostate carcinoma xenografts, and Clare Lamont and Kristin Gullage for figure graphics.


    REFERENCES
 Top
 Notes
 Introduction
 Microvessel Density Is A...
 Microvessel Density May Not...
 Looking Beyond Measurements of...
 References
 

1 Brem S, Cotran R, Folkman J. Tumor angiogenesis: a quantitative method for histologic grading. J Natl Cancer Inst 1972;48:347–56.[Medline]

2 Weidner N, Semple JP, Welch WR, Folkman J. Tumor angiogenesis and metastasis—correlation in invasive breast carcinoma. N Engl J Med 1991;324:1–8.[Abstract]

3 Weidner N, Folkman J, Pozza F, Bevilacqua P, Allred EN, Moore DH, et al. Tumor angiogenesis: a new significant and independent prognostic indicator in early-stage breast carcinoma. J Natl Cancer Inst 1992;84:1875–87.[Abstract]

4 Weidner N, Carroll PR, Flax J, Blumenfeld W, Folkman J. Tumor angiogenesis correlates with metastasis in invasive prostate carcinoma. Am J Pathol 1993;143:401–9.[Abstract]

5 de Jong JS, van Diest PJ, Baak JP. Hot spot microvessel density and the mitotic activity index are strong additional prognostic indicators in invasive breast cancer. Histopathology 2000;36:306–12.[Medline]

6 Borre M, Offersen BV, Nerstrom B, Overgaard J. Microvessel density predicts survival in prostate cancer patients subjected to watchful waiting. Br J Cancer 1998;78:940–4.[Medline]

7 Yano T, Tanikawa S, Fujie T, Masutani M, Horie T. Vascular endothelial growth factor expression and neovascularisation in non-small cell lung cancer. Eur J Cancer 2000;36:601–9.[Medline]

8 Tanaka F, Otake Y, Yanagihara K, Kawano Y, Miyahara R, Li M, et al. Evaluation of angiogenesis in non-small cell lung cancer: comparison between anti-CD34 antibody and anti-CD105 antibody. Clin Cancer Res 2001;7:3410–5.[Abstract/Free Full Text]

9 de la Taille A, Katz AE, Bagiella E, Buttyan R, Sharir S, Olsson CA, et al. Microvessel density as a predictor of PSA recurrence after radical prostatectomy. A comparison of CD34 and CD31. Am J Clin Pathol 2000;113:555–62.[Medline]

10 Ushijima C, Tsukamoto S, Yamazaki K, Yoshino I, Sugio K, Sugimachi K. High vascularity in the peripheral region of non-small cell lung cancer tissue is associated with tumor progression. Lung Cancer 2001;34:233–41.[Medline]

11 Gasparini G. Clinical significance of determination of surrogate markers of angiogenesis in breast cancer. Crit Rev Oncol Hematol 2001;37:97–114.[Medline]

12 Gasparini G, Harris AL. Prognostic significance of tumor vascularity. In: Teicher BA, editor. Antiangiogenic agents in cancer therapy. Totowa (NJ): Humana Press; 1999. p. 317–99.

13 Srivastava A, Laidler P, Davies RP, Horgan K, Hughes LE. The prognostic significance of tumor vascularity in intermediate-thickness (0.76–4.0 mm thick) skin melanoma. A quantitative histologic study. Am J Pathol 1988;133:419–23.[Abstract]

14 Fallowfield ME, Cook MG. The vascularity of primary cutaneous melanoma. J Pathol 1991;164:241–4.[Medline]

15 Carnochan P, Briggs JC, Westbury G, Davies AJ. The vascularity of cutaneous melanoma: a quantitative histological study of lesions 0.85–1.25 mm in thickness. Br J Cancer 1991;64:102–7.[Medline]

16 Busam KJ, Berwick M, Blessing K, Fandrey K, Kang S, Karaoli T, et al. Tumor vascularity is not a prognostic factor for malignant melanoma of the skin. Am J Pathol 1995;147:1049–56.[Abstract]

17 Graham CH, Rivers J, Kerbel RS, Stankiewicz KS, White WL. Extent of vascularization as a prognostic indicator in thin (<0.76 mm) malignant melanomas. Am J Pathol 1994;145:510–4.[Abstract]

18 Vlaykova T, Muhonen T, Hahka-Kemppinen M, Pyrhonen S, Jekunen A. Vascularity and prognosis of metastatic melanoma. Int J Cancer 1997;74:326–9.[Medline]

19 Makitie T, Summanen P, Tarkkanen A, Kivela T. Microvascular density in predicting survival of patients with choroidal and ciliary body melanoma. Invest Ophthalmol Vis Sci 1999;40:2471–80.[Abstract/Free Full Text]

20 Macchiarini P, Fontanini G, Hardin MJ, Squartini F, Angeletti CA. Relation of neovascularisation to metastasis of non-small-cell lung cancer. Lancet 1992;340:145–6.[Medline]

21 Yamazaki K, Abe S, Takekawa H, Sukoh N, Watanabe N, Ogura S, et al. Tumor angiogenesis in human lung adenocarcinoma. Cancer 1994;74:2245–50.[Medline]

22 Fontanini G, Bigini D, Vignati S, Basolo F, Mussi A, Lucchi M, et al. Microvessel count predicts metastatic disease and survival in non-small cell lung cancer. J Pathol 1995;177:57–63.[Medline]

23 Giatromanolaki A, Koukourakis M, O'Byrne K, Fox S, Whitehouse R, Talbot DC, et al. Prognostic value of angiogenesis in operable non-small cell lung cancer. J Pathol 1996;179:80–8.[Medline]

24 Angeletti CA, Lucchi M, Fontanini G, Mussi A, Chella A, Ribechini A, et al. Prognostic significance of tumoral angiogenesis in completely resected late stage lung carcinoma (stage IIIA-N2). Impact of adjuvant therapies in a subset of patients at high risk of recurrence. Cancer 1996;78:409–15.[Medline]

25 Apolinario RM, van der Valk P, de Jong JS, Deville W, van Ark-Otte J, Dingemans AM, et al. Prognostic value of the expression of p53, bcl-2, and bax oncoproteins, and neovascularization in patients with radically resected non-small-cell lung cancer. J Clin Oncol 1997;15:2456–66.[Abstract]

26 Dazzi C, Cariello A, Maioli P, Solaini L, Scarpi E, Rosti G, et al. Prognostic and predictive value of intratumoral microvessels density in operable non-small-cell lung cancer. Lung Cancer 1999;24:81–8.[Medline]

27 Han H, Silverman JF, Santucci TS, Macherey RS, d'Amato TA, Tung MY, et al. Vascular endothelial growth factor expression in stage I non-small cell lung cancer correlates with neoangiogenesis and a poor prognosis. Ann Surg Oncol 2001;8:72–9.[Abstract/Free Full Text]

28 Cox G, Walker RA, Andi A, Steward WP, O'Byrne KJ. Prognostic significance of platelet and microvessel counts in operable non-small cell lung cancer. Lung Cancer 2000;29:169–77.[Medline]

29 O'Byrne KJ, Koukourakis MI, Giatromanolaki A, Cox G, Turley H, Steward WP, et al. Vascular endothelial growth factor, platelet-derived endothelial cell growth factor and angiogenesis in non-small-cell lung cancer. Br J Cancer 2000;82:1427–32.[Medline]

30 Marrogi AJ, Travis WD, Welsh JA, Khan MA, Rahim H, Tazelaar H, et al. Nitric oxide synthase, cyclooxygenase 2, and vascular endothelial growth factor in the angiogenesis of non-small cell lung carcinoma. Clin Cancer Res 2000;6:4739–44.[Abstract/Free Full Text]

31 Aikawa H, Takahashi H, Fujimura S, Sato M, Endo C, Sakurada A, et al. Immunohistochemical study on tumor angiogenic factors in non-small cell lung cancer. Anticancer Res 1999;19:4305–9.[Medline]

32 Masuya D, Huang C, Liu D, Kameyama K, Hayashi E, Yamauchi A, et al. The intratumoral expression of vascular endothelial growth factor and interleukin-8 associated with angiogenesis in nonsmall cell lung carcinoma patients. Cancer 2001;92:2628–38.[Medline]

33 Fontanini G, Lucchi M, Vignati S, Mussi A, Ciardiello F, De Laurentiis M, et al. Angiogenesis as a prognostic indicator of survival in non-small-cell lung carcinoma: a prospective study. J Natl Cancer Inst 1997;89:881–6.[Abstract/Free Full Text]

34 Fontanini G, Vignati S, Lucchi M, Mussi A, Calcinai A, Boldrini L, et al. Neoangiogenesis and p53 protein in lung cancer: their prognostic role and their relation with vascular endothelial growth factor (VEGF) expression. Br J Cancer 1997;75:1295–301.[Medline]

35 Olivarez D, Ulbright T, DeRiese W, Foster R, Reister T, Einhorn L, et al. Neovascularization in clinical stage A testicular germ cell tumor: prediction of metastatic disease. Cancer Res 1994;54:2800–2.[Abstract]

36 Jones A, Fujiyama C, Turner K, Fuggle S, Cranston D, Turley H, et al. Angiogenesis and lymphangiogenesis in stage 1 germ cell tumours of the testis. BJU Int 2000;86:80–6.

37 West CM, Cooper RA, Loncaster JA, Wilks DP, Bromley M. Tumor vascularity: a histological measure of angiogenesis and hypoxia. Cancer Res 2001;61:2907–10.[Abstract/Free Full Text]

38 Wakui S, Furusato M, Itoh T, Sasaki H, Akiyama A, Kinoshita I, et al. Tumour angiogenesis in prostatic carcinoma with and without bone marrow metastasis: a morphometric study. J Pathol 1992;168:257–62.[Medline]

39 Fregene TA, Khanuja PS, Noto AC, Gehani SK, Van Egmont EM, Luz DA, et al. Tumor-associated angiogenesis in prostate cancer. Anticancer Res 1993;13:2377–81.[Medline]

40 Vesalainen S, Lipponen P, Talja M, Alhava E, Syrjanen K. Tumor vascularity and basement membrane structure as prognostic factors in T1–2MO prostatic adenocarcinoma. Anticancer Res 1994;14: 709–14.[Medline]

41 Brawer MK, Deering RE, Brown M, Preston SD, Bigler SA. Predictors of pathologic stage in prostatic carcinoma. The role of neovascularity. Cancer 1994;73:678–87.[Medline]

42 Silberman MA, Partin AW, Veltri RW, Epstein JI. Tumor angiogenesis correlates with progression after radical prostatectomy but not with pathologic stage in Gleason sum 5 to 7 adenocarcinoma of the prostate. Cancer 1997;79:772–9.[Medline]

43 Barth PJ, Weingartner K, Kohler HH, Bittinger A. Assessment of the vascularization in prostatic carcinoma: a morphometric investigation. Hum Pathol 1996;27:1306–10.[Medline]

44 Rogatsch H, Hittmair A, Reissigl A, Mikuz G, Feichtinger H. Microvessel density in core biopsies of prostatic adenocarcinoma: a stage predictor? J Pathol 1997;182:205–10.[Medline]

45 Halvorsen OJ, Haukaas S, Hoisaeter PA, Akslen LA. Independent prognostic importance of microvessel density in clinically localized prostate cancer. Anticancer Res 2000;20:3791–9.[Medline]

46 Rubin MA, Buyyounouski M, Bagiella E, Sharir S, Neugut A, Benson M, et al. Microvessel density in prostate cancer: lack of correlation with tumor grade, pathologic stage, and clinical outcome. Urology 1999;53:542–7.[Medline]

47 Dickinson AJ, Fox SB, Persad RA, Hollyer J, Sibley GN, Harris AL. Quantification of angiogenesis as an independent predictor of prognosis in invasive bladder carcinomas. Br J Urol 1994;74:762–6.[Medline]

48 Bochner BH, Cote RJ, Weidner N, Groshen S, Chen SC, Skinner DG, et al. Angiogenesis in bladder cancer: relationship between microvessel density and tumor prognosis. J Natl Cancer Inst 1995;87:1603–12.[Abstract]

49 Grossfeld GD, Ginsberg DA, Stein JP, Bochner BH, Esrig D, Groshen S, et al. Thrombospondin-1 expression in bladder cancer: association with p53 alterations, tumor angiogenesis, and tumor progression. J Natl Cancer Inst 1997;89:219–27.[Abstract/Free Full Text]

50 Korkolopoulou P, Konstantinidou AE, Kavantzas N, Patsouris E, Pavlopoulos PM, Christodoulou P, et al. Morphometric microvascular characteristics predict prognosis in superficial and invasive bladder cancer. Virchows Arch 2001;438:603–11.[Medline]

51 Reiher F, Ozer O, Pins M, Jovanovic BD, Eggener S, Campbell SC. p53 and microvessel density in primary resection specimens of superficial bladder cancer. J Urol 2002;167:1469–74.[Medline]

52 Tanigawa N, Matsumura M, Amaya H, Kitaoka A, Shimomatsuya T, Lu C, et al. Tumor vascularity correlates with the prognosis of patients with esophageal squamous cell carcinoma. Cancer 1997;79:220–5.[Medline]

53 Shih CH, Ozawa S, Ando N, Ueda M, Kitajima M. Vascular endothelial growth factor expression predicts outcome and lymph node metastasis in squamous cell carcinoma of the esophagus. Clin Cancer Res 2000;6:1161–8.[Abstract/Free Full Text]

54 Hironaka S, Hasebe T, Kamijo T, Ohtsu A, Boku N, Yoshida S, et al. Biopsy specimen microvessel density is a useful prognostic marker in patients with T(2–4)M(0) esophageal cancer treated with chemoradiotherapy. Clin Cancer Res 2002;8:124–30.[Abstract/Free Full Text]

55 Maeda K, Chung YS, Takatsuka S, Ogawa Y, Sawada T, Yamashita Y, et al. Tumor angiogenesis as a predictor of recurrence in gastric carcinoma. J Clin Oncol 1995;13:477–81.[Abstract]

56 Tanigawa N, Amaya H, Matsumura M, Shimomatsuya T. Association of tumour vasculature with tumour progression and overall survival of patients with non-early gastric carcinomas. Br J Cancer 1997;75:566–71.[Medline]

57 Sanz-Ortega J, Steinberg SM, Moro E, Saez M, Lopez JA, Sierra E, et al. Comparative study of tumor angiogenesis and immunohistochemistry for p53, c-ErbB2, c-myc and EGFr as prognostic factors in gastric cancer. Histol Histopathol 2000;15:455–62.[Medline]

58 Saclarides TJ, Speziale NJ, Drab E, Szeluga DJ, Rubin DB. Tumor angiogenesis and rectal carcinoma. Dis Colon Rectum 1994;37:921–6.[Medline]

59 Tomisaki S, Ohno S, Ichiyoshi Y, Kuwano H, Maehara Y, Sugimachi K. Microvessel quantification and its possible relation with liver metastasis in colorectal cancer. Cancer 1996;77(8 Suppl):1722–8.[Medline]

60 Takebayashi Y, Aklyama S, Yamada K, Akiba S, Aikou T. Angiogenesis as an unfavorable prognostic factor in human colorectal carcinoma. Cancer 1996;78:226–31.[Medline]

61 Bossi P, Viale G, Lee AK, Alfano R, Coggi G, Bosari S. Angiogenesis in colorectal tumors: microvessel quantitation in adenomas and carcinomas with clinicopathological correlations. Cancer Res 1995;55:5049–53.[Abstract]

62 Cianchi F, Palomba A, Messerini L, Boddi V, Asirelli G, Perigli G, et al. Tumor angiogenesis in lymph node-negative rectal cancer: correlation with clinicopathological parameters and prognosis. Ann Surg Oncol 2002;9:20–6.[Abstract/Free Full Text]

63 Pietra N, Sarli L, Caruana P, Cabras A, Costi R, Gobbi S, et al. Is tumour angiogenesis a prognostic factor in patients with colorectal cancer and no involved nodes? Eur J Surg 2000;166:552–6.[Medline]

64 Sternfeld T, Foss HD, Kruschewski M, Runkel N. The prognostic significance of tumor vascularization in patients with localized colorectal cancer. Int J Colorectal Dis 1999;14:272–6.[Medline]

65 Lindmark G, Gerdin B, Sundberg C, Pahlman L, Bergstrom R, Glimelius B. Prognostic significance of the microvascular count in colorectal cancer. J Clin Oncol 1996;14:461–6.[Abstract]

66 Rajkumar SV, Mesa RA, Tefferi A. A review of angiogenesis and anti-angiogenic therapy in hematologic malignancies. J Hematother Stem Cell Res 2002;11:33–47.[Medline]

67 Sezer O, Niemoller K, Eucker J, Jakob C, Kaufmann O, Zavrski I, et al. Bone marrow microvessel density is a prognostic factor for survival in patients with multiple myeloma. Ann Hematol 2000;79:574–7.[Medline]

68 Munshi NC, Wilson C. Increased bone marrow microvessel density in newly diagnosed multiple myeloma carries a poor prognosis. Semin Oncol 2001;28:565–9.[Medline]

69 Di Raimondo F, Palumbo GA, Molica S, Giustolisi R. Angiogenesis in chronic myeloproliferative diseases. Acta Haematol 2001;106: 177–83.[Medline]

70 Moehler TM, Neben K, Ho AD, Goldschmidt H. Angiogenesis in hematologic malignancies. Ann Hematol 2001;80:695–705.[Medline]

71 Rajkumar SV, Kyle RA. Angiogenesis in multiple myeloma. Semin Oncol 2001;28:560–4.[Medline]

72 Folkman J. What is the evidence that tumors are angiogenesis dependent? J Natl Cancer Inst 1990;82:4–6.[Medline]

73 Rak J, Filmus J, Kerbel RS. Reciprocal paracrine interactions between tumour cells and endothelial cells: the `angiogenesis progression' hypothesis. Eur J Cancer 1996;32A:2438–50.

74 Weidner N. Tumour vascularity and proliferation: clear evidence of a close relationship. J Pathol 1999;189:297–9.[Medline]

75 Perez-Atayde AR, Sallan SE, Tedrow U, Connors S, Allred E, Folkman J. Spectrum of tumor angiogenesis in the bone marrow of children with acute lymphoblastic leukemia. Am J Pathol 1997;150:815–21.[Abstract]

76 Thomlinson RH, Gray LH. The histological structure of some human lung cancers and the possible implications for radiotherapy. Br J Cancer 1955;9:539–49.

77 Steinberg F, Rohrborn HJ, Otto T, Scheufler KM, Streffer C. NIR reflection measurements of hemoglobin and cytochrome aa3 in healthy tissue and tumors. Correlations to oxygen consumption: preclinical and clinical data. Adv Exp Med Biol 1997;428:69–77.[Medline]

78 Eberhard A, Kahlert S, Goede V, Hemmerlein B, Plate KH, Augustin HG. Heterogeneity of angiogenesis and blood vessel maturation in human tumors: implications for antiangiogenic tumor therapies. Cancer Res 2000;60:1388–93.[Abstract/Free Full Text]

79 Turner HE, Nagy Z, Gatter KC, Esiri MM, Harris AL, Wass JA. Angiogenesis in pituitary adenomas and the normal pituitary gland. J Clin Endocrinol Metab 2000;85:1159–62.[Abstract/Free Full Text]

80 Strohmeyer D, Rossing C, Strauss F, Bauerfeind A, Kaufmann O, Loening S. Tumor angiogenesis is associated with progression after radical prostatectomy in pT2/pT3 prostate cancer. Prostate 2000;42:26–33.[Medline]

81 Hanahan D, Weinberg RA. The hallmarks of cancer. Cell 2000;100:57–70.[Medline]

82 Bartelink H, Begg A, Martin JC, van Dijk M, van't Veer L, van der Vaart P, et al. Towards prediction and modulation of treatment response. Radiother Oncol 1999;50:1–11.[Medline]

83 Crompton NE, Ozsahin M, Schweizer P, Larsson B, Luetolf UM. Theory and practice of predictive assays in radiation therapy. Strahlenther Onkol 1997;173:58–67.[Medline]

84 Chapman JD, Engelhardt EL, Stobbe CC, Schneider RF, Hanks GE. Measuring hypoxia and predicting tumor radioresistance with nuclear medicine assays. Radiother Oncol 1998;46:229–37.[Medline]

85 Wesseling P, van der Laak JA, Link M, Teepen HL, Ruiter DJ. Quantitative analysis of microvascular changes in diffuse astrocytic neoplasms with increasing grade of malignancy. Hum Pathol 1998;29:352–8.[Medline]

86 Beecken WD, Fernandez A, Joussen AM, Achilles EG, Flynn E, Lo KM, et al. Effect of antiangiogenic therapy on slowly growing, poorly vascularized tumors in mice. J Natl Cancer Inst 2001;93:382–7.[Abstract/Free Full Text]

87 Folkman J, Hahnfeldt P, Hlatky L. Cancer: looking outside the genome. Nat Rev Mol Cell Biol 2000;1:76–9.[Medline]

88 Relf M, LeJeune S, Scott PA, Fox S, Smith K, Leek R, et al. Expression of the angiogenic factors vascular endothelial cell growth factor, acidic and basic fibroblast growth factor, tumor growth factor beta-1, platelet-derived endothelial cell growth factor, placenta growth factor, and pleiotrophin in human primary breast cancer and its relation to angiogenesis. Cancer Res 1997;57:963–9.[Abstract]

89 Hahnfeldt P, Panigrahy D, Folkman J, Hlatky L. Tumor development under angiogenic signaling: a dynamical theory of tumor growth, treatment response, and postvascular dormancy. Cancer Res 1999;59:4770–5.[Abstract/Free Full Text]

90 Hlatky L, Tsionou C, Hahnfeldt P, Coleman CN. Mammary fibroblasts may influence breast tumor angiogenesis via hypoxia-induced vascular endothelial growth factor up-regulation and protein expression. Cancer Res 1994;54:6083–6.[Abstract]

91 Igarashi M, Dhar DK, Kubota H, Yamamoto A, El-Assal O, Nagasue N. The prognostic significance of microvessel density and thymidine phosphorylase expression in squamous cell carcinoma of the esophagus. Cancer 1998;82:1225–32.[Medline]

92 Smith K, Fox SB, Whitehouse R, Taylor M, Greenall M, Clarke J, et al. Upregulation of basic fibroblast growth factor in breast carcinoma and its relationship to vascular density, oestrogen receptor, epidermal growth factor receptor and survival. Ann Oncol 1999;10:707–13.[Abstract]

93 Kodama J, Seki N, Tokumo K, Hongo A, Miyagi Y, Yoshinouchi M, et al. Platelet-derived endothelial cell growth factor is not implicated in progression of cervical cancer. Oncol Rep 1999;6:617–20.[Medline]

94 Terris B, Scoazec JY, Rubbia L, Bregeaud L, Pepper MS, Ruszniewski P, et al. Expression of vascular endothelial growth factor in digestive neuroendocrine tumours. Histopathology 1998;32:133–8.[Medline]

95 Yu JL, Rak JW, Coomber BL, Hicklin DJ, Kerbel RS. Effect of p53 status on tumor response to antiangiogenic therapy. Science 2002;295:1526–8.[Abstract/Free Full Text]

96 Freemantle SJ, Dmitrovsky E. Clinical link between p53 and angiogenesis in lung cancer. J Clin Oncol 2002;20:883–4.[Free Full Text]

97 Vaupel P, Hoeckel M. Predictive power of the tumor oxygenation status. Adv Exp Med Biol 1999;471:533–9.[Medline]

98 Shima DT, Adamis AP, Ferrara N, Yeo KT, Yeo TK, Allende R, et al. Hypoxic induction of endothelial cell growth factors in retinal cells: identification and characterization of vascular endothelial growth factor (VEGF) as the mitogen. Mol Med 1995;1:182–93.[Medline]

99 Stein I, Neeman M, Shweiki D, Itin A, Keshet E. Stabilization of vascular endothelial growth factor mRNA by hypoxia and hypoglycemia and coregulation with other ischemia-induced genes. Mol Cell Biol 1995;15:5363–8.[Abstract]

100 Brown L, Detmar M, Claffey K, Nagy J, Feng D, Dvorak A, et al. Vascular permeability factor/vascular endothelial growth factor: a multifunctional angiogenic cytokine. In: Goldberg ID, Rosen EM, editors. Regulation of angiogenesis. Boston (MA): Birkhauser; 1997.

101 Dvorak HF, Brown LF, Detmar M, Dvorak AM. Vascular permeability factor/vascular endothelial growth factor, microvascular hyperpermeability, and angiogenesis. Am J Pathol 1995;146:1029–39.[Abstract]

102 Ravi R, Mookerjee B, Bhujwalla ZM, Sutter CH, Artemov D, Zeng Q, et al. Regulation of tumor angiogenesis by p53-induced degradation of hypoxia-inducible factor 1alpha. Genes Dev 2000;14:34–44.[Abstract/Free Full Text]

103 Kondo K, Kaelin WG Jr. The von Hippel-Lindau tumor suppressor gene. Exp Cell Res 2001;264:117–25.[Medline]

104 Graeber TG, Osmanian C, Jacks T, Housman DE, Koch CJ, Lowe SW, et al. Hypoxia-mediated selection of cells with diminished apoptotic potential in solid tumours. Nature 1996;379:88–91.[Medline]

105 Pluda JM. Tumor-associated angiogenesis: mechanisms, clinical implications, and therapeutic strategies. Semin Oncol 1997;24:203–18.[Medline]

106 Herbst RS, Hidalgo M, Pierson AS, Holden SN, Bergen M, Eckhardt SG. Angiogenesis inhibitors in clinical development for lung cancer. Semin Oncol 2002;29(1 Suppl 4):66–77.[Medline]

107 Singhal S, Mehta J, Desikan R, Ayers D, Roberson P, Eddlemon P, et al. Antitumor activity of thalidomide in refractory multiple myeloma. N Engl J Med 1999;341:1565–71.[Abstract/Free Full Text]

108 Raje N, Anderson K. Thalidomide—a revival story. N Engl J Med 1999;341:1606–9.[Free Full Text]

109 Bertolini F, Mingrone W, Alietti A, Ferrucci PF, Cocorocchio E, Peccatori F, et al. Thalidomide in multiple myeloma, myelodysplastic syndromes and histiocytosis. Analysis of clinical results and of surrogate angiogenesis markers. Ann Oncol 2001;12:987–90.[Abstract]

110 Stoler DL, Chen N, Basik M, Kahlenberg MS, Rodriguez-Bigas MA, Petrelli NJ, et al. The onset and extent of genomic instability in sporadic colorectal tumor progression. Proc Natl Acad Sci U S A 1999;96:15121–6.[Abstract/Free Full Text]

111 O'Reilly MS, Holmgren L, Shing Y, Chen C, Rosenthal RA, Moses M, et al. Angiostatin: a novel angiogenesis inhibitor that mediates the suppression of metastases by a Lewis lung carcinoma. Cell 1994;79:315–28.[Medline]

112 Boehm T, Folkman J, Browder T, O'Reilly MS. Antiangiogenic therapy of experimental cancer does not induce acquired drug resistance. Nature 1997;390:404–7.[Medline]

113 Anderson GR, Stoler DL, Brenner BM. Cancer: the evolved consequence of a destabilized genome. Bioessays 2001;23:1037–46.[Medline]

114 Benjamin LE, Hemo I, Keshet E. A plasticity window for blood vessel remodelling is defined by pericyte coverage of the preformed endothelial network and is regulated by PDGF-B and VEGF. Development 1998;125:1591–8.[Abstract/Free Full Text]

115 Benjamin LE, Golijanin D, Itin A, Pode D, Keshet E. Selective ablation of immature blood vessels in established human tumors follows vascular endothelial growth factor withdrawal. J Clin Invest 1999;103:159–65.[Abstract/Free Full Text]

116 Goede V, Schmidt T, Kimmina S, Kozian D, Augustin HG. Analysis of blood vessel maturation processes during cyclic ovarian angiogenesis. Lab Invest 1998;78:1385–94.[Medline]

117 Kakolyris S, Fox SB, Koukourakis M, Giatromanolaki A, Brown N, Leek RD, et al. Relationship of vascular maturation in breast cancer blood vessels to vascular density and metastasis, assessed by expression of a novel basement membrane component, LH39. Br J Cancer 2000;82:844–51.[Medline]

118 St Croix B, Rago C, Velculescu V, Traverso G, Romans KE, Montgomery E, et al. Genes expressed in human tumor endothelium. Science 2000;289:1197–202.[Abstract/Free Full Text]

119 Veikkola T, Alitalo K. VEGFs, receptors and angiogenesis. Semin Cancer Biol 1999;9:211–20.[Medline]

120 Gale NW, Yancopoulos GD. Growth factors acting via endothelial cell-specific receptor tyrosine kinases: VEGFs, angiopoietins, and ephrins in vascular development. Genes Dev 1999;13:1055–66.[Free Full Text]

121 Carmeliet P. Mechanisms of angiogenesis and arteriogenesis. Nat Med 2000;6:389–95.[Medline]

122 Auerbach W, Auerbach R. Angiogenesis inhibition: a review. Pharmacol Ther 1994;63:265–311.[Medline]

123 Mauceri HJ, Hanna NN, Beckett MA, Gorski DH, Staba MJ, Stellato KA, et al. Combined effects of angiostatin and ionizing radiation in antitumour therapy. Nature 1998;394:287–91.[Medline]

124 Gorski DH, Mauceri HJ, Salloum RM, Gately S, Hellman S, Beckett MA, et al. Potentiation of the antitumor effect of ionizing radiation by brief concomitant exposures to angiostatin. Cancer Res 1998;58:5686–9.[Abstract]

125 Browder T, Butterfield CE, Kraling BM, Shi B, Marshall B, O'Reilly MS, et al. Antiangiogenic scheduling of chemotherapy improves efficacy against experimental drug-resistant cancer. Cancer Res 2000;60:1878–86.[Abstract/Free Full Text]

126 Klement G, Baruchel S, Rak J, Man S, Clark K, Hicklin DJ, et al. Continuous low-dose therapy with vinblastine and VEGF receptor-2 antibody induces sustained tumor regression without overt toxicity. J Clin Invest 2000;105:R15–24.[Medline]

127 Drixler TA, Rinkes IH, Ritchie ED, van Vroonhoven TJ, Gebbink MF, Voest EE. Continuous administration of angiostatin inhibits accelerated growth of colorectal liver metastases after partial hepatectomy. Cancer Res 2000;60:1761–5.[Abstract/Free Full Text]

128 Hahnfeldt P, Hlatky L. Cell resensitization during protracted dosing of heterogeneous cell populations. Radiat Res 1998;150:681–7.[Medline]

129 Mross K. Anti-angiogenesis therapy: concepts and importance of dosing schedules in clinical trials. Drug Resist Updat 2000;3:223–35.[Medline]

130 Bergers G, Javaherian K, Lo KM, Folkman J, Hanahan D. Effects of angiogenesis inhibitors on multistage carcinogenesis in mice. Science 1999;284:808–12.[Abstract/Free Full Text]

131 Hanahan D, Bergers G, Bergsland E. Less is more, regularly: metronomic dosing of cytotoxic drugs can target tumor angiogenesis in mice. J Clin Invest 2000;105:1045–7.[Free Full Text]

132 Gasparini G. Metronomic scheduling: the future of chemotherapy? Lancet Oncol 2001;2:733–40.[Medline]

133 Fidler IJ, Ellis LM. Chemotherapeutic drugs—more really is not better. Nat Med 2000;6:500–2.[Medline]

134 Kamen BA, Rubin E, Aisner J, Glatstein E. High-time chemotherapy or high time for low dose. J Clin Oncol 2000;18:2935–7.[Free Full Text]

135 Gately S, Kerbel R. Antiangiogenic scheduling of lower dose cancer chemotherapy. Cancer J 2001;7:427–36.[Medline]

136 Soffer SZ, Kim E, Moore JT, Huang J, Yokoi A, Manley C, et al. Novel use of an established agent: Topotecan is anti-angiogenic in experimental Wilms tumor. J Pediatr Surg 2001;36:1781–4.[Medline]

Manuscript received September 11, 2001; revised April 5, 2002; accepted April 11, 2002.


This article has been cited by other articles in HighWire Press-hosted journals:


             
Copyright © 2002 Oxford University Press (unless otherwise stated)
Oxford University Press Privacy Policy and Legal Statement