anti-angiogenic drugs: from bench to clinical trials

48
Anti-Angiogenic Drugs: from Bench to Clinical Trials Ana R. Quesada, 1 Ramo´n Mun˜oz-Cha´puli, 2 Miguel A. Medina 1 1 Department of Molecular Biology and Biochemistry, Faculty of Science, University of Ma ´ laga, 29071 Ma ´ laga, Spain 2 Department of Animal Biology, Faculty of Science, University of Ma ´ laga, 29071 Ma ´ laga, Spain Published online 1 May 2006 in Wiley InterScience (www.interscience.wiley.com). DOI 10.1002/med.20059 ! Abstract: Angiogenesis, the generation of new capillaries through a process of pre-existing microvessel sprouting, is under stringent control and normally occurs only during embryonic and post-embryonic development, reproductive cycle, and wound repair. However, in many pathological conditions (solid tumor progression, metastasis, diabetic retinopathy, hemangioma, arthritis, psoriasis and atherosclerosis among others), the disease appears to be associated with persistent upregulated angiogenesis. The development of specific anti-angiogenic agents arises as an attractive therapeutic approach for the treatment of cancer and other angiogenesis-dependent diseases. The formation of new blood vessels is a complex multi-step process. Endothelial cells resting in the parent vessels are activated by an angiogenic signal and stimulated to synthesize and release degradative enzymes allowing endothelial cells to migrate, proliferate and finally differentiate to give rise to capillary tubules. Any of these steps may be a potential target for pharmacological intervention. In spite of the disappointing results obtained initially in clinical trials with anti-angiogenic drugs, recent reports with positive results in phases II and III trials encourage expectations in their therapeutic potential. This review discusses the current approaches for the discovery of new compounds that inhibit angiogenesis, with emphasis on the clinical developmental status of anti-angiogenic drugs. ß 2006 Wiley Periodicals, Inc. Med Res Rev, 26, No. 4, 483–530, 2006 Key words: angiogenesis; inhibitors; cancer; macular degeneration; clinical trials 1. INTRODUCTION TO ANTI-ANGIOGENIC THERAPY Angiogenesis, the formation of new blood vessels from the existing vascular bed, has been described as one of the hallmarks of cancer, playing an essential role in tumor growth, invasion, and metastasis. Correspondence to: Dr. Ana R. Quesada, Department of Molecular Biology and Biochemistry, Faculty of Science, University of Ma¤ laga, E-29071Ma¤ laga, Spain. E-mail: [email protected] Medicinal Research Reviews, Vol. 26, No. 4, 483 ^530, 2006 ß 2006 Wiley Periodicals, Inc.

Upload: independent

Post on 07-Feb-2023

0 views

Category:

Documents


0 download

TRANSCRIPT

Anti-Angiogenic Drugs:from Bench to ClinicalTrials

Ana R. Quesada,1 Ramon Munoz-Chapuli,2 Miguel A. Medina1

1Department of Molecular Biology and Biochemistry, Faculty of Science, University of Malaga,

29071 Malaga, Spain2Department of Animal Biology, Faculty of Science, University of Malaga,

29071 Malaga, Spain

Published online 1 May 2006 in Wiley InterScience (www.interscience.wiley.com).

DOI 10.1002/med.20059

!

Abstract: Angiogenesis, the generation of new capillaries through a process of pre-existing

microvessel sprouting, is under stringent control and normally occurs only during embryonic and

post-embryonic development, reproductive cycle, and wound repair. However, in many

pathological conditions (solid tumor progression, metastasis, diabetic retinopathy, hemangioma,

arthritis, psoriasis and atherosclerosis among others), the disease appears to be associated with

persistent upregulated angiogenesis. The development of specific anti-angiogenic agents arises as

an attractive therapeutic approach for the treatment of cancer and other angiogenesis-dependent

diseases. The formation of new blood vessels is a complex multi-step process. Endothelial cells

resting in the parent vessels are activated by an angiogenic signal and stimulated to synthesize

and release degradative enzymes allowing endothelial cells to migrate, proliferate and finally

differentiate to give rise to capillary tubules. Any of these steps may be a potential target for

pharmacological intervention. In spite of the disappointing results obtained initially in clinical

trials with anti-angiogenic drugs, recent reports with positive results in phases II and III trials

encourage expectations in their therapeutic potential. This review discusses the current approaches

for the discovery of new compounds that inhibit angiogenesis, with emphasis on the clinical

developmental status of anti-angiogenic drugs. � 2006 Wiley Periodicals, Inc. Med Res Rev, 26, No. 4,

483–530, 2006

Key words: angiogenesis; inhibitors; cancer; macular degeneration; clinical trials

1 . I N T R O D U C T I O N T O A N T I - A N G I O G E N I C T H E R A P Y

Angiogenesis, the formation of new blood vessels from the existing vascular bed, has been described

as one of the hallmarks of cancer, playing an essential role in tumor growth, invasion, andmetastasis.

Correspondence to: Dr. Ana R.Quesada, Department of Molecular Biology and Biochemistry, Faculty of Science, University of

Ma¤ laga,E-29071Ma¤ laga,Spain.E-mail: [email protected]

Medicinal Research Reviews, Vol. 26, No. 4, 483^530, 2006

� 2006 Wiley Periodicals, Inc.

Furthermore, many other diseases are also dependent on upregulated angiogenesis. When resting

endothelial cells are activated by an angiogenic signal, they are stimulated to release degrading

enzymes allowing endothelial cells to migrate, proliferate, and finally differentiate to form new

vessels. Figure 1 is a scheme of the angiogenic process.Anyof the steps involved in angiogenesismay

be a potential target for pharmacological intervention of angiogenesis-dependent diseases. This is the

main reasonwhy angiogenesis has attracted recent attention in the field of pharmacological research.

Anti-angiogenic therapy, directed to activated endothelial cells, presents a series of advantages

when compared to traditional therapies, mainly targeted to tumor cells. First, endothelial cells are

homogeneous, diploid, genetically stable targets. Spontaneous mutations occur very rarely in them.

This is in contrast to the high genetic instability, heterogeneity, and mutation rate of tumor cells,

which favors the selection of subpopulations that become resistant to the anti-tumoral drugs, resulting

in the appearance of chemotherapy resistance phenomena. Second, the turnover of endothelial cells

in tumoral tissues may be 50-fold higher than that of quiescent endothelia in normal tissues. The

activated endothelium overexpresses specific markers, such as integrin avb3, E-selectin, Tie, andvascular endothelial growth factor (VEGF) receptors that could be used for the development of

specific therapies directed to these cells. On the other hand, the contact between endothelial cells and

blood makes this target easily accessible to systemically administered drugs, therefore avoiding the

problem of low penetration of the anti-tumoral drugs into the solid tumors.1 Finally, due to the fact

that a singlevascular netmay support the growth of different populations of tumor cells, the inhibition

of the vascular growth may affect the survival of many tumor cells.2

There are different strategies for angiogenesis intervention, based on the modulation of any of

the different steps of the angiogenic process (Fig. 2). Therefore, some angiogenesis modulators may

Figure 1. Scheme of the steps of the angiogenic process. An activating, proangiogenic signal1elicits the switching on of the

angiogenic phenotype of resting endothelial cells, which become activated.2These activated cells acquire an invasive potential,

involving basal membrane degradation,3extracellular matrix remodeling, proliferation, and migration.

4Finally, morphogenesis

contributes to thealignmentofendothelial cells forminganewmicrovessel, and this is stabilizedbyeventual recruitmentofpericytes

andacquisitionofanewbasalmembrane.5

484 * QUESADA, MUN‹ OZ-CHA¤ PULI, AND MEDINA

act on the angiogenic signal, or on one or several specific functions of activated endothelial cells

(proliferation, adhesion to extracellular matrix, protease production, migration, invasion or

differentiation). In this article, the main angiogenesis inhibitors will be classified according to the

step of angiogenesis they affect.

Figure 2. Thereare different strategies to inhibitangiogenesis, basedoneffectiveandspecificblockageofanyof the different stepsof the angiogenic process. Accordingly, anti-angiogenic drugs can be classified in the several groups described in the present

work, includingdrugs inhibiting the angiogenic factor (AF) andone or several functions ofactivated endothelial cells (proliferation,

adhesion, proteaseproduction, migration, invasion, anddifferentiation).

ANTI-ANGIOGENIC DRUGS * 485

2 . A G E N T S T H A T I N T E R F E R E W I T H T H E A N G I O G E N I C S I G N A L

Among the growth factors that are involved in the angiogenesis process, VEGF and basic fibroblast

growth factor (bFGF) seem to play the most relevant roles. For this reason, a number of therapeutic

strategies are aimed at neutralizing the activation of endothelial cells by these factors. The following

is a brief summary of such studies.

A. Agents Acting on the Angiogenic Factor

The production of neutralizing antibodies anti-VEGF stands out. One specific and prominent

example is the humanized recombinant monoclonal antibody rhu Anti-VEGF Mab (Bevacizumab,

Avastin, developed by Genentech, South San Francisco, CA; www.genentech.com). This drug was

generated by engineering the VEGF-binding residues of a murine neutralizing antibody into the

framework of a normal human immunoglobulin G (IgG). The humanization provides a longer half-

life and less immunogenicity. Bevacizumab neutralizes the biologically active forms of VEGF that

interact with VEGF receptors 1 and 2 (VEGFR1 andVEGFR2), which results in the neutralization of

the biological properties of human VEGF, including endothelial cell mitogenic activity, vascular-

permeability-enhancing activity, and angiogenic properties.3 The anti-tumor activity of this antibody

has been extensively demonstrated in pre-clinical studies in several tumor models. These studies

found that single-agent therapy with bevacizumab or its murine equivalent (A4.6.1) resulted in tumor

growth inhibition of a number of human tumor cell lines xenografted in rodentmodels, irrespective of

the route of administration or tumor location. The assayed tumor types included rhabdomyosarcoma,

glioblastoma, leiomyosarcoma, colon adenocarcinoma, hepatoblastoma, neuroblastoma, Wilms’

tumor, and ovarian, prostate or breast carcinomas.4 Studies with in vitro cell lines did not reveal a

direct effect of the antibody on the growth of tumor cells, suggesting that tumor suppression is

mediated through inhibition of neovascularization.5 Several studies have shown that anti-VEGF

therapy also prevented the growth of tumor cells at metastatic sites.4

Clinical data obtained to date indicate that most patients tolerated bevacizumab well when

administered alone or in combination with the traditional chemotherapy, although serious adverse

events have been observed. Side effects can include high blood pressure, nosebleeds, fever, headache,

rash, and chills. Occasionally, bevacizumab treatment could be associated with blood clots and

bleeding,which could be life threatening. From a safety point of view, themechanismof bleeding and

thrombosis associated with bevacizumab treatment remains to be clarified.6 Bevacizumab is being

evaluated in combination with standard therapies in phases II and III clinical trials of colorectal

cancer (CRC), renal cell carcinoma (RCC), breast cancer, Kaposi’s sarcoma, pancreatic cancer,

ovarian cancer, lymphoma, myeloma, melanoma, head and neck cancer, leukemia, liver cancer,

sarcoma, mesothelioma, and lung cancer. The most encouraging results have been obtained in the

treatment of metastatic CRC, RCC, breast cancer, and non-small cell lung cancer (NSCLC) patients.

The possibility of using bevacizumab in combination with chemo or radiotherapy seems very

promising,7 as suggested by results obtained in the treatment of advanced colon cancer.8 In a

randomized, open-label phase II assay, carried out with 104 metastatic cancer patients, subjects with

previously untreated metastatic cancer, were treated with a combination of bevacizumab and the

traditional chemotherapy (5-FU/LV). The results indicate that this combination therapy was clearly

superior to those obtainedwith chemotherapy alone. The first study showing phase III data validating

an anti-angiogenesis strategy for treating human cancers was presented in a communication that

received a lot of attention in the 2003American Society of Clinical Oncology (ASCO)Meeting.9 The

authors found that the bevacizumab/IFL (irinotecan/5FU/LV) combination led to significantly

prolonged survival. It also had a better ability to shrink tumors than standard IFL alone, without

statistically significant increases in adverse events in patients with metastatic colorectal cancer. The

results were based on 412 patients in the IFL/placebo arm and 403 patients in the IFL/bevacizumab

486 * QUESADA, MUN‹ OZ-CHA¤ PULI, AND MEDINA

arm. The presence of bevacizumab in the treatment produced remarkable and statistically very

significant increases in all the four survival and response parameters determined: median survival,

progression-free survival, objective response, and duration of response.10 Based on these data,

bevacizumab received the US Food and Drug Administration (FDA) approval in 2004 for use in

combination with fluorouracil-based chemotherapy as first-line treatment for metastatic cancer of

the colon or rectum. Bevacizumab has also received a positive recommendation from the European

Committee for Medicinal Products for Human Use in 2004. In early 2005, preliminary results

from a large, randomized clinical trial for patients with advanced colorectal cancer who had

previously received treatment were made public. Those results indicate that bevacizumab

administered in combination with oxaliplatin-based chemotherapy (FOLFOX4) is well tolerated

and increases survival in advanced CRC patients previously treated with irinotecan and a

fluoropyrimidine.11,12

Bevacizumab has also been recently tested in metastatic renal cancer, a tumor with high

expression of VEGF resulting from inactivation of the von Hippel-Lindau (VHL) tumor suppressor

gene.13 In a randomized, double-blind, phase II trial therewas a significant prolongation of the time to

progression of disease in the high-dose-antibody group as compared with the placebo group

(4.8 months vs. 2.5 months).14 Recent studies in combination with the standard treatment, interferon

(IFN), or with erlotinib, a low molecular weight inhibitor of the endothelial growth factor receptor

(EGFR) tyrosin kinase are generating promising data in RCC treatment. In a phase II trial including

58 patients with recurrent RCC that underwent nephrectomy, 87% of patients treated with erlotinib/

bevacizumab achieved a clinical benefit, with 21% of patients achieving at least a 50% reduction in

their cancer size and 66% showing either a disease stabilization of aminor anti-cancer response. Only

13% of patients had a progression of their cancer while on treatment. One year later, 81% of patients

were still alive. These rates are some of the highest rates ever achieved in patients with this stage of

RCC. Side effects caused by treatment were generally mild, with the most common side effects

being high blood pressure, diarrhea, rash, nausea, and vomiting.15,16 An Intergroup phase III trial

investigating the addition of bevacizumab to initial systemic therapy in RCC is being evaluated to

see whether the addition of anti-VEGF therapy to IFN alpha would prolong survival in untreated

metastatic RCC patients.17

A randomized phase III trial comparing the efficacy and safety of capecitabine with or without

bevacizumab, in patients withmetastatic breast cancer previously treatedwith an anthracycline and a

taxane failed to show improvement in survival in bevacizumab treated patients.18 However, in April

2005 a press release from Genentech reported an interim analysis of a phase III study in first-line

metastatic breast cancer, showing bevacizumab plus paclitaxel improved progression-free survival

when compared with chemotherapy alone. This trial enrolled 722 women with previously untreated

metastatic breast cancer, who were randomized to receive paclitaxel with or without bevacizumab.

An additional ‘late-breaking’ session was created in the 41st Annual Meeting of the American

Society of Clinical Oncology (Orlando, FL, May 2005) to accommodate the presentation of these

results. Data presented at ASCO 2005, based on 355 events observed, showed that progression-free

survival (primary endpoint) was 6.11 months for paclitaxel and 10.97 months for the combination

arm, for a hazard ratio of 0.498 (95% CI, 0.401–0.618), and a P-value of <0.001. A trend for

improvement in overall survival was also noted, with a hazard ratio of 0.674 (95%CI, 0.495–0.917),

P ¼ 0.01. In terms of toxicity, both regimens were well tolerated. This is the first time an anti-

angiogenic agent has been found to be successful in metastatic breast cancer.19

In the same way, promising results have been recently reported for the treatment of lung cancer

patients with bevacizumab in combination with standard chemotherapy or erlotinib.20,21 A large,

randomized phase II/III clinical trial sponsored by the USNational Cancer Institute (NCI) focuses in

patients with previously untreated advanced non-squamous, NSCLC. Preliminary results from this

trial show that adding bevacizumab to paclitaxel and carbiplatin compared with paclitaxel/

carboplatin by itself improved survival 2 months.22

ANTI-ANGIOGENIC DRUGS * 487

The use of soluble receptors is a different strategy to sequester the angiogenic factor. The

production of a soluble form of the receptor VEGFR1 by human endothelial and tumor cells has been

described, as well as its presence in plasma and serum from healthy donors.23 These observations

suggest that these soluble receptors may play a physiological role in the regulation of the biological

activity of VEGF. As an application of this principle to the pharmacological modulation of

angiogenesis, it has been reported that the use of extracellular domains of VEGFR1, VEGFR2, and

Tie-2 potently inhibited corneal neovascularization induced by tumor cell conditioned media, and

reduced mammary tumor growth and vascularization when they were administered into a rat

cutaneous tumor window chamber.24,25 Based on the results obtained with cancer cells infected with

adenovirus vectors encoding soluble VEGFR1, it has been suggested that anti-angiogenic gene

therapy using soluble VEGFR1 might be an effective approach for pancreatic or lung cancer

treatment. The tumor growth of the immuno-depressedmice thatwere inoculatedwith the genetically

modified cancer cells was significantly suppressed. The microvessel density in this group was also

significantly lower.26,27 Moreover, when adenovirus expressing the entire extracellular domain of

VEGFR1 was injected into the mice skeletal muscle and uninfected lung cancer cells were injected

s.c., the soluble receptor was detectable in the circulating blood. This was accompanied by the

cessation of tumor growth, and later a decrease of tumor size. Histological examination of the animal

models revealed that intratumor angiogenesis was markedly suppressed, and apoptosis was

enhanced. These results suggest that adenovirus-mediated overexpression of a soluble VEGF

receptor in a remote organ may have the potential to be an effective strategy for cancer treatment.27

The success of this experimental strategy can be helped by the possible synergy between VEGF and

bFGF soluble receptors, as well as their possible production in remote organs, therefore maintaining

their anti-angiogenic activity.27,28 Soluble forms of VEGFR1 suffer from poor pharmacokinetic

properties, which seem to correlate with their non-specific interactions with extracellular matrix.

By determining the requirements to maintain high affinity while extending in vivo half-life, a

very strong high-affinity VEGF blocker that has markedly enhanced pharmacokinetic properties

has been engineered. The VEGF-Trap (Aventis and Regeneron Pharmaceuticals, Tarrytown, NY

www.regeneron.com) is a derivative of VEGFR1, comprising portions of the extracellular domains

of both VEGFR1 and VEGFR2 fused to an Fc segment of IgG and therefore minimizing the

possibility of being immunogenic in humans. The combination of high affinity and improved

pharmacokinetics apparently contributes toward making VEGF-Trap one of the most potent and

efficacious VEGF-blockers available, showing picomolar affinity for both mouse and human

VEGF.29 Systemic administration of VEGF-Trap dramatically inhibited the growth and

vascularization of a variety of tumor cell lines, including mouse melanoma, human rhabdomyo-

sarcoma, and rat glioma.29 In a xenografted model of neuroblastoma, treatment with high-dose

VEGF-Trap resulted in stunted tumors, which were virtually avascular and in which only rare co-

opted glomeruli were present, suggesting that the co-opted vasculature had regressed.30 Remarkably,

VEGF blockade resulted in dramatic remodeling of the blood vessels in disseminated ovarian

carcinoma, prevented ascites accumulation, and inhibited the growth of disseminated cancer.31 Some

data suggest that blockade of VEGF may not only impede cancer progression, presumably by

preventing neoangiogenesis, but could also produce tumor regression. It has been reported that

high-affinity blockade of VEGF, using the VEGF-Trap, abolishes mature, pre-existing vasculature

in established xenografts. Eradication of vasculature is followed by marked tumor regression,

including that of lung micrometastases. These results support the importance of VEGF as a target in

cancer therapy and suggest that anti-VEGF strategies may not only halt tumor growth but also

produce actual regression. Such results also suggest that potent blockade ofVEGFmay be effective in

the treatment of patients with metastatic, bulky cancers, as well as those with minimal residual

disease.32 On the other hand, VEGF-Trapmay also provide a new agent for treatment of patients with

choroidal neovascularization and diabetic macular edema, since VEGF plays a central role in the

development of both diseases.33 VEGF-Trap is being evaluated in phase I in patients with solid

488 * QUESADA, MUN‹ OZ-CHA¤ PULI, AND MEDINA

tumors and non-Hodgkin’s lymphomas, and for the treatment of age-related macular degeneration

(AMD).34

A different approach for therapeutic inhibition of angiogenesis employs drugs that inhibit the

angiogenic factor production. Taking this into account, it has been reported that interferon alpha

(IFN-a) (Schering Corp., Kenilworth, NJ, www.schering.com) decreases the expression of bFGF,

very widespread in hemangiomas,35 and inhibits VEGF transcription through an Sp1-transcription

and/or Sp3-dependent inhibition of VEGF promoter activity.36 IFN-a has been generally well

tolerated by children suffering from severe hemangiomas, and has given positive results in the

treatment of hemangiomas and other childhood tumors, as well as in metastatic melanoma

patients.37–48 The combination of temozolomide/IFN proved quite tolerable in a phase II trial

evaluating their efficacy in patients with advanced renal cell carcinoma. This regime appeared

inactive in terms of response in this population with poor prognosis. It was more promising in those

patients with diseases remaining stable for more than 6 months.49 The use of aggressive supportive

measures and appropriate dose-reduction strategies may lessen the toxicity of high doses of IFN.50

IFN-a is being evaluated in phase III for renal cell carcinoma, squamous cell carcinoma of the head

and neck, and in phase II for unresectable carcinoid tumors, multiple myeloma, metastatic

melanoma, and high grade gliomas. There are more than 60 clinical studies with IFN-a under course.Since bFGF biological activity depends on extracellular heparin concentration, bFGF activity

can be modulated in vivo by the use of heparin analogs. Among others, suramin, a polysulphonated

naphtylurea first synthesized as an agent against trypanosomiasis and onchocerciasis, should be

mentioned. Suramin was found to inhibit the binding of a number of peptide growth factors such as

PDGF and bFGF to their receptors by binding to the growth factor and by inducing a conformational

change and therefore inhibiting their biological signal.51–53 Suramin interacts with VEGFR2 and is a

potent VEGF antagonist.54 In vitro, suramin inhibited multiple control points of angiogenesis,

including endothelial cell migration, proliferation, and proteolytic activities.55 In vivo, suramin

inhibited angiogenesis in a number of experimental models, including the chick chorioallantoic

membrane (CAM) assay, the rat corneal and rat aortic ring assays, and gelatin sponges loaded with

bFGF and implanted subcutaneously in mice.55–57 Suramin reduced tumor growth and tumor

induced angiogenesis in the murine M5076 reticulosarcoma model, a tumor producing significant

levels of bFGF.57 Suramin has been extensively evaluated over the last 15 years as an anti-cancer

agent, with the most interest being the treatment of prostate cancer. Early clinical results were

promising with response rates of up to 70% being reported. However, a recent double-blind study

showed only modest palliative effect in patients with androgen-independent prostate cancer. In

retrospect, it appears that those initial reports failed to control for confounding variables such as anti-

androgen withdrawal and hydrocortisone.58 The clinical use of suramin is being complicated by a

broad spectrum of toxic effects, neurotoxicity being the most significant complication and appearing

to be related to the intensity of the dosage.59–61 Although suramin clinical use is limited by the

appearance of the mentioned toxicities, an improvement in the treatment regime has produced

enhanced response with decreased toxicity in a phase II trial in 58 patients with hormone refractory

prostate cancer.62 Suramin is currently being evaluated in phase I in patients with NSCLC or bladder

cancer and in phase II in patients with NSCLC or breast cancer.

Suramin toxicity could be overcome by the synthesis of less toxic compounds that maintain

suramin anti-angiogenic properties.63–65 Suradista NSC 651016 is a distamicine sulfonated

derivative of suramin that is also effective at complexing and inactivating growth factors and

cytokines while remaining relatively non-toxic. NSC 651016 is more effective than suramin in

inhibiting mitogenesis in normal or transformed cells, as well as the growth of xenografts grown in

athymic nude mice.66 NSC 651016 acts as an inhibitor of stromal cell-derived factor 1 alpha

(CXCL12)-mediated angiogenesis in a variety of in vitro and in vivo angiogenesis assays, including

endothelial cell migration, capillary-like tube formation, aortic ring, and matrigel plug assay.67 NSC

651016 has been evaluated in phase I for solid tumors and in phase II for recurrent primary brain

ANTI-ANGIOGENIC DRUGS * 489

tumors. The promising anti-angiogenic properties of NSC 651016 evaluated with serum bFGF as the

surrogate end point have not been achievable with i.v. administration in patients with solid tumors.68

Pentosan polysulfate (PPS) is a semi-synthetic pentasaccharide heparinoid derived from beech

wood shavings that has been used as anti-coagulant for more than 30 years. PPS is an effective

inhibitor of heparin-binding growth factors in vitro and can effectively inhibit the establishment and

growth of tumors in nude mice. Pentosan inhibited the growth of rat prostate adenocarcinoma when

treatment was started when the tumor was not palpable, but had little effect against established

tumors. PPS inhibited endothelial cell motility and tubule formation in vitro as well as capillary

formation in the CAM assay.69 PPS has been evaluated in phase I/II in advanced cancer and Kaposi’s

sarcoma, but its clinical application has been limited by the high anti-coagulant activity at doses

needed to inhibit the angiogenesis.70,71 A mild response has been reported in a phase II clinical trial

with patients with AIDS-related Kaposi’s sarcoma treated with PPS.72 New interest may arise from

its use in the treatment of inflammatory diseases and osteoarthritis.73,74

Finally, some attempts are being devoted to the development of chimerical molecules where

toxins are bound to VEGF or their receptors. Targeted toxins, developed by recombinant methods by

fusing VEGF165 or VEGF121 to the diphtheria toxin were found to be highly toxic to proliferating

endothelial cells but not to vascular smooth muscle cells. These fusion proteins completely inhibited

the bFGF-induced growth of new blood vessels in the CAM assay. Furthermore, the fusion toxin

substantially retarded the growth of Kaposi’s sarcoma tumors in mice.75 Dividing endothelial cells

overexpressing VEGFR2 have been reported to be approximately 60-fold more sensitive to a fusion

protein containing VEGF121 linked to the toxin gelonin. Endothelial cells overexpressing VEGFR1

were not sensitive to this fusion protein. Treatment of humanmelanomaor human prostate xenografts

with the fusion construct demonstrated a reduction in tumor volume and caused thrombotic damage

to tumor vessels with extravasation of red blood cells in the tumor bed.76 Whereas these preliminary

studies show a potential use of fusion protein in a variety of disorders in which angiogenesis plays a

role, certain limitations are inherent to the use of bacterial proteins due to the generation of antibodies

and the appearance of non-targeted toxicities. The development of less immunogenic immunotoxins

could permit repetitive dosing, improving their therapeutic performance.77

B. Agents Acting on the Receptor of the Angiogenic Factor

Some authors think that inhibiting VEGF receptors may be more effective than blocking their

ligands, sincemany cellular types can express VEGF, and this factor can be stored in the extracellular

matrix and in the tissular interstitial spaces. VEGF levels may therefore be very variable in space and

time, and this makes its neutralization difficult. On the contrary, the number of VEGF receptors in the

activated endothelial cells is relatively low, and, at least in theory, more easily saturable, since less

antagonist molecules would be needed to block all the receptors.78

Away to inhibit the expression of a receptor is by means of a specific ribozyme. A ribozyme is a

catalytic RNA able to hydrolyze a given RNA. Its specificity is derived from the matching between

the ribozyme binding arms, and the sequences flanking the cleavage site in the target RNA.

Ribozymes for clinical use are designed to resist in tissues and biological fluids that are rich in

nucleases. They have an adequate pharmacokinetic profile and have demonstrated their effectiveness

in in vitro and in vivo pre-clinical studies.79 Significant inhibition of angiogenesis using ribozymes

against both VEGFR1 and VEGFR2 has been reported. In animal tumor models, anti-tumor effects

were most pronounced with the anti-VEGFR1 ribozyme known as Angiozyme (Ribozyme

Pharmaceuticals, Boulder, CO, www.rpi.com). Angiozyme has demonstrated anti-angiogenic

effects in a rat cornea model of VEGF-induced angiogenesis and anti-tumor and anti-metastatic

activity in several murine tumor models.80–82 Extensive pre-clinical studies in mice and monkeys

have demonstrated no significant toxicities of angiozyme.83,84 Angiozyme has beenwell tolerated by

humans in phase I clinical studies, and has been evaluated in phase II trials in advanced and/or

490 * QUESADA, MUN‹ OZ-CHA¤ PULI, AND MEDINA

metastatic cancer patients alone or in combination with traditional chemotherapy.85–87 However,

regardless of the phase II trial results, the company has announced that it does not intend to develop

angiozyme independently. This compound might be licensed to a third party partner, if found.

In a less advanced stage of development, we could mention the possible use of peptides to

interfere the binding site of VEGF and its receptor. These studies will throw light on the interactions

of growth factor and their receptor.88,89

Another experimental strategy to pharmacologic intervention of angiogenesis consists of

receptor blockade by antibodies generated against the receptor extracellular domain. The use of

monoclonal antibodies generated against the different VEGF receptors inhibits tumor angiogenesis

and human tumor growth in immunodepressed mice.3,90–92 The humanized anti-VEGFR2 IMC-

1C11 antibodies (Imclone, New York, NY, www.imclone.com) bind specifically to the endothelial

cell surface extracellular domain of VEGFR2, blocks its interaction with VEGF, and prevents VEGF

activation of the intracellular tyrosine kinase pathway.93 IMC-1C11 inhibits VEGF-stimulated

proliferation of human endothelial cells and migration of VEGFR2þ leukemia cells in vitro. When

administered in vivo, these antibodies significantly prolonged survival of NOD-SCID mice

inoculated with VEGFR2þ human leukemia cells. IMC-1C11 has been well tolerated in phase I in

patients suffering from colorectal carcinomawith livermetastasis, but no objective tumor regressions

were observed.92,94 IMC-1121B is a fully human monoclonal antibody that targets VEGFR2,

produced as a second-generation agent, which is expected to be non-immunogenic for chronic

administration as a single agent and in combination with chemotherapy or radiation. IMC-1121B

binds to VEGFR2 with high affinity and efficiently blocks its interaction with VEGF. IMC-1121B

also inhibits VEGF-stimulated VEGFR2 activation and endothelial cell migration and prolifera-

tion.95,96 In January 2005, Im Clone Systems has initiated a phase I clinical trial of IMC-1121B in

patients with solid tumors.

The angiogenesis inhibitory activities of antibodies anti-VEGFR1 and anti-VEGFR3 are

currently being studied, but they have not reached the clinical phases of development yet.97 A

bispecific antibody, a diabody, using the variable domain genes of IMC-1121B (specific to VEGFR2)

and hF4-3C5 (specific to VEGFR3) as the ‘‘building blocks,’’ has recently been constructed. The

diabody binds to both VEGFR2 and VEGFR3 in a dose-dependent manner and blocks interaction

between these receptors and VEGF. In cell-based assays, the diabody neutralized both VEGF-

stimulated endothelial cell migration and activation of the receptors and p42/p44 mitogen-activated

protein kinases (MAPK) in endothelial cells. These results suggest that a simultaneous dual blockade

of both VEGFR2 and VEGFR3 may represent a more potent approach to effective cancer therapy.98

C. Agents Interfering the Signaling Pathway of the Angiogenic Factor

As mentioned above, there is no doubt about the main role played by VEGF and its receptors in the

angiogenic process. For this reason, a number of efforts have been devoted to the search of drugs that

are able to inhibit the VEGF receptors signaling pathways, and in particular their autophos-

phorylation.

Among the compounds that inhibit VEGFR2we can remark SU5416 and SU6668 (Fig. 3), which

have received a high clinical interest. Both compounds compete with ATP for the receptor tyrosine

kinase domain.99 In addition, SU5416 (Semaxanib, SUGEN/Pharmacia, South San Francisco, CA,

www.sugen.com) inhibits tyrosine kinase activity of PDGF receptors, VEGFR1, VEGFR3, and c-

kit.78 The first results obtainedwith this compound in vitro and in vivowere impressive, showing anti-

tumoral activity even when the drug was administered once the tumor had reached a size that was

detectable in the animal. SU5416 has been shown to inhibit VEGF-dependent mitogenesis of human

endothelial cellswithout inhibiting the growth of a variety of tumor cells invitro. In contrast, systemic

administration of SU5416 at non-toxic doses in mice resulted in inhibition of subcutaneous tumor

growth of cells derived from various tissue origins.99 SU5416 inhibited metastases, microvessel

ANTI-ANGIOGENIC DRUGS * 491

formation, and increased tumor and endothelial cell apoptosis in mice injected with CT-26 colon

cancer cells to generate metastases.100 Reduction in tumor growth in a human neurogenic sarcoma

explant xenograft model was due to decreased tumor angiogenesis, leading to reduction of tumor cell

proliferation and increased apoptosis.101 SU5416, when combined with irradiation, has an additive

effect over treatment with irradiation alone in a human glioblastoma xenograft. Permanent vascular

changes were seen after combined treatment resulting in complete remission.102 Its long lasting

effects in vivo suggested the possibility of a weekly administration in the clinical assays.103

Unfortunately, the results arising from the clinical studies have not confirmed these expectations. The

use of SU5416 in phases I and II clinical studies for the treatment of sarcoma,melanoma, von-Hippel

Lindau disease, and head and neck, breast, ovary, prostate andmetastatic renal cancers has given only

partially positive results in some cases.104–112 Phase III assays of co-administration of SU5416,

5-FU, and LV tometastatic colon cancer patients were a failure, casting doubts on the design of these

clinical trials. Among other undesired effects, risk of thromboembolisms after the SU5416

administration should be mentioned.113 All of these problems caused the total interruption of the

mentioned clinical trial in 2002. Lately, a phase II study conducted to investigate the safety and

efficacy of SU5416 for patients with advanced soft tissue sarcomas, has shown that this agent was

relativelywell tolerated, but did not demonstrate significant anti-tumor activity.114 Phase II studies of

single agent SU5416 showed a modest clinical activity in patients with refractory acute myeloid

leukemia (AML) or myelodysplastic syndromes, and in patients with refractory multiple

myeloma.115,116 Although prolonged remission was observed in a few patients with c-kit-positive

AML that was resistant to standard chemotherapy, the overall observed response rate was low and

responses consisted mainly of partial remissions of short duration.117 No clinical benefit was shown

in phase II studies in patients with RCC, advanced soft tissue sarcoma, advanced melanoma or

hormone refractory prostate cancer.118–121

Figure 3. Inhibitors of theactivationofangiogenic factor receptors.

492 * QUESADA, MUN‹ OZ-CHA¤ PULI, AND MEDINA

SU6668 (SUGEN, Inc., co-developed with Taiho Pharmaceutical Co., Tokyo, Japan) is another

VEGFR2 inhibitor, which also inhibits bFGF and PDGF receptors.122 SU6668 has competitive

inhibitory properties with respect to ATP, inhibits phosphotyrosine levels in VEGFR2 and platelet-

derived growth factor receptor (PDGFR) b in vivo, and rapidly induces tumormicro vessel apoptosis,

followed by apoptosis of tumor cells.123 In cellular systems, it inhibits receptor tyrosine

phosphorylation and mitogenesis in a dose-dependent manner after stimulation of cells by

appropriate ligands. Oral or i.p. administration of SU6668 in athymic mice resulted in significant

growth inhibition of a diverse panel of human tumor xenografts of glioma, melanoma, lung, colon,

ovarian, and epidermoid origin. Furthermore, intravital multi-fluorescence video microscopy of C6

glioma xenografts in the dorsal skinfold chamber model revealed that SU6668 treatment suppressed

tumor angiogenesis. SU6668 treatment induced striking regression of large established human tumor

xenografts.124 SU6668 increased median survival by 58% (P < 0.001). It also led to a progressive

increase in tumor cell and endothelial cell apoptosis that increased over time in BALB/c mice that

underwent splenic injectionwith CT-26 colon cancer cells to generate livermetastases.125 SU6668 in

combination with paclitaxel inhibited ovarian carcinoma progression in the peritoneal cavity, by

blocking ascites formation and tumor spread, in ovarian carcinoma xenograft models transplanted in

the peritoneal cavity of nude mice.126 The simultaneous inhibition of several receptors that are

involved in the angiogenic response of endothelial cells might increase the possibilities for the

pharmacological development of this compound. SU6668 induced a tumoral regression in lung and

colon experimental tumors, even when the treatment was initiated when the tumor mass was

considerably high.122,123 SU6668 has been assayed in phase I/II clinical studies with advanced

tumors and multiple myeloma patients, but no objective responses have been observed.126–128 A

recent study about the effects of SU5416 and SU6668 on receptor phosphorylation in tumor

xenografts and in paired tumor biopsies obtained in three clinical trials in patients with advanced

solid malignancies showed that both compounds displayed biological activity in xenografts.

However, neither drug produced marked biological activity in patients with primary tumors.129

Outstanding interest has been reached by the development of the new compound SU11248

(Fig. 3) (Sutent, sunitinib malate, Pfizer, Inc., NewYork, NY, www.pfizer.com), that is able to inhibit

the tyrosine kinase activity of a number of receptors including PDGFR, VEGFR2, Flt-3, and c-Kit.

SU11248 can be administered orally and exhibits an interesting anti-tumoral activity, as shown in

pre-clinical studies. SU11248 exhibited broad and potent anti-tumor activity causing regression,

growth arrest, or substantially reduced growth of various established mouse xenografts derived

from human or rat tumor cell lines.130 SU11248 dramatically regressed FLT3-ITD tumors in

the subcutaneous tumor xenograft model and prolonged survival in the bone marrow engraft-

ment model.131 SU11248 treatment potently regressed growth of mammary cancers in mouse

mammary tumor virus-v-Ha-ras transgenic mice and 7,12-dimethylbenz(a)anthracene-induced

mammary tumors in rats. This agent also inhibited tumor growth in human breast cancer xenografts of

s.c. MX-1 tumors. SU11248 treatment in combination with docetaxel effectively prolonged survival

of mice.132 It has been suggested that SU11248 might be an effective and tolerated therapy to inhibit

growth of breast cancer bone metastases, with the additional advantage of inhibiting tumor-

associated osteolysis.133 SU11248 enhanced radiation-induced endothelial cytotoxicity, resulting in

tumor vascular destruction and tumor control when combined with fractionated radiotherapy in

murine tumor models. A synergistic interaction of SU11248 and the traditional anti-leukemic agents

cytarabine or daunorubicin in leukemia cells has been reported.134,135 Recent results from a phase I

clinical study show that SU11248 monotherapy induced partial remissions of short duration in AML

patients.136

In a phase II clinical trial involving 63 renal cancer patients who had already tried IL-2 or IFN

without success, as a result a 40% had a partial response. Tumors did not progress for more than 3

months in an additional 28% of patients, indicating that 68% of patients benefited from SU11248

treatment. In addition, the average time to tumor progression for patients in this studywas 8.7months,

ANTI-ANGIOGENIC DRUGS * 493

and the median overall survival was 16.4 months. A second phase II study of 106 patients

demonstrated an objective response rate of 39% in patients treatedwith SU11248. In addition, 23%of

patients experienced tumor stabilization. Taken together, a total of 62% of patients benefited from

treatment with SU11248.137–139 Results from these two studies suggest that SU11248 has substantial

anti-tumor activity inmetastatic renal cell carcinoma as second-line therapy and form the basis for the

launch of a large-scale phase III program to determine the potential benefits of SU11248 in earlier

stage disease.

A phase I study in Gastrointestinal Stromal Tumor (GIST), which had stopped responding to

other treatment gave encouraging results. In a phase I/II study, SU11248 therapy induced objective

responses in patients with several mutational variants of GIST displaying different genomic

mechanisms of resistance to imatinib mesylate (IM). Long-term follow-up data from this study have

demonstrated that SU11248 extended overall survival to nearly 20 months in patients whose cancer

had progressed despite treatment with other standard therapies. In addition, the median time to tumor

progression in this study was 7.8 months for all patients, with some specific subtypes of patients

benefiting even more dramatically than would be expected with IM.140,141 These data were the basis

for a phase III trial to further define the activity of SU11248 in patients with IM-resistant GIST. In

early 2005, Pfizer reported that this phase III randomized study has successfully met its efficacy

endpoint. Results from this double-blind phase III study of more than 300 patients showed SU11248

significantly prolonged the time to tumor progression (6.3 months on SU11248 vs. 1.5 months for

controls) and reduced the risk of death by approximately 50% compared to placebo.142These results

substantiate the concept that multi-targeted molecular therapy can overcome resistance to other

targeted drugs in cancer. SU11248 is currently being tested in phase II clinical trials in patients with

previously treated metastatic breast cancer and in patients with unresectable neuroendocrine

tumors.143,144

PTK787/ZK222584 (Fig. 3) (vatalanib, Novartis Pharma AG, Basel, Switzerland, www.

novartis.com, co-developed with Schering AG, Berlin, www.schering.de) is an orally available

aminophthalazine derivative that strongly inhibits all known VEGFR tyrosine kinases expressed on

endothelial cells. It also inhibits other class III kinases, such as the PDGFR beta tyrosine kinase, c-

Kit, and c-Fms, but at higher concentrations. It is not active against kinases from other receptor

families, such as EGFR, FGFR1, c-Met, and Tie-2, or intracellular kinases such as c-Src, c-Abl, and

protein kinase C-a. The compound inhibits the endothelial cell proliferation, migration, and survival

in the nanomolar range in cell-based assays. After oral administration, it induced a dose-dependent

inhibition of VEGF and PDGF-induced angiogenesis in a growth factor implant model, and it

inhibited the growth of several human carcinomas, grown s.c. in nude mice, as well as a murine renal

carcinoma and its metastases in a syngeneic, orthotopic model. It did not affect wound healing.145

Oral administration of PTK787 alone or in combination with gemcitabine decreased the growth and

metastasis of human pancreatic carcinoma growing orthotopically in nude mice.146,147 Although the

clinical development of this drug was initially interrupted because of the appearance of some

undesired toxicities, it was reinitiated after checking that those toxicities were reversible and of

benign nature. In various phase I studies, daily oral administration was well tolerated.148,149 A phase

I/II study has shown that PTK787 can be safely combined with FOLFOX4 (oxaliplatin, fluorouracil,

folinic acid) in patients with metastatic CRC. At the higher doses of PTK787, dizziness and

neurologic toxicities were seen.150 Ongoing randomized phase III trials (CONFIRM-1 and

CONFIRM-2) are evaluating the efficacy and safety of PTK787 in combination with FOLFOX4 in

patients with newly diagnosed or relapsed advanced stage CRC.151 In March 2005, the company

announced that the analysis of CONFIRM-1 results indicated that pre-planned analysis of

progression-free survival achieved statistical significance. At the same time, analysis of primary

endpoint of progression-free survival assessed by central review did not achieve statistical

significance. Independent data monitoring board has recommended CONFIRM-1 trial to continue to

allow analysis of overall survival (Second Primary Endpoint). Novartis and Schering have now

494 * QUESADA, MUN‹ OZ-CHA¤ PULI, AND MEDINA

anticipated filing for approval with the US FDA and the European Medicines Agency (EMEA) in

early 2007. Moreover, encouraging anti-tumor activity was seen with PTK787 monotherapy as well

as in combination with temozolomide or lomustine in a phase I/II study in patients with recurrent

glioblastoma multiforme.152 PTK787 is also currently being evaluated in Phase II studies for the

treatment of myelodysplastic syndromes, malignant mesothelioma, and von Hippel-Lindau disease.

Von Hippel-Lindau disease (VHL) is a familiar hereditary cancer caused by mutations in the VHL

suppressor gene germ line. It is characterized by the appearance of very vascularized tumors,

overexpressing VEGF, in the retina and in the central nervous system. For these reasons, they are

good candidates for the anti-angiogenic therapies directed to block VEGF action.

The drug ZD4190 (Astra Zeneca Pharmaceuticals LP, Macclesfield, Cheshire, UK, www.

astrazeneca.com) is a 4-anilinoquinazoline (Fig. 3) that inhibits the tyrosine kinase activities of

VEGFR2 andVEGFR1, as well as theVEGF stimulated human endothelial cell proliferation in vitro.

It shows anti-tumoral activity in breast, lung, prostate, and ovarian human tumor xenografts; upon

withdrawal of therapy, tumor growth resumed after a short delay.153 ZD6474 (Fig. 3) is a soluble

anilinoquinazoline that selectively inhibits the VEGFR2 tyrosine kinase and inhibits, to a lesser

extent, the activity of other tyrosine kinases such as those of the EGFR and the PDGFR. ZD6474

blocks VEGF-stimulated endothelial cell migration and proliferation. Chronic, daily oral

administration of ZD6474 produced significant broad-spectrum anti-tumor activity on human tumor

xenografts implanted in nude mice.154 It has also been reported that ZD6474 has the potential to

enhance the effect of radiotherapy and potentiates the anti-tumoral activity of taxanes in vitro and

in vivo.155,156 Recent studies have demonstrated that the inhibition of EGFR phosphorylation by

ZD6474 can contribute significantly to the anti-tumor efficacy of this agent.157 Phase I clinical

evaluation has shown that ZD6474 is generally well tolerated, and tumor responses in NSCLC have

been documented.158,159 Phase II clinical studies of ZD6474 oral administration to patients of lung

cancer or multiple myeloma are in progress.160,161

Sorafenib (BAY 43-9006, Bayer Pharmaceuticals Corporation, Leverkusen, Germany, www.

pharma.bayer.com, andOnyxPharmaceuticals, Inc., Emerville, CA,www.onyx-pharm.com) is a novel

bi-aryl urea that inhibits tumor cell proliferation and tumor growth in several human tumor xenograft

models by targeting the RAF/MEK/EKR signaling pathway at the level of RAF kinase.162,163 In

addition, BAY 43-9006 demonstrated significant activity against several receptor tyrosine kinases

involved in neovascularization and tumor progression, including VEGFR2, VEGFR3, PDGFR beta,

Flt-3, and c-KIT. Once daily oral dosing of sorafenib demonstrated broad-spectrum anti-tumor

activity in colon, breast, and NSCLC xenograft models. Analyses of microvessel density and

microvessel area in the same tumor sections using anti-murine CD31 antibodies demonstrated

significant inhibition of neovascularization in all three of the xenograft models.164

Sorafenib has beenwell tolerated and showed anti-tumoral activity in phase I studies in advanced

refractory solid tumors.165–168 A phase II randomized discontinuation study showed a significantly

better progression-free survival rate with sorafenib in renal cancer.169–171 In May 2005, Bayer and

Onyx announced that an ongoing multi-national, phase III study in patients with RCC or kidney

cancer had met its surrogate endpoint, resulting in statistically significant longer progression-free

survival in those patients administered with sorafenib. More than 900 patients, who had previously

failed one prior systemic therapy, were randomized one-to-one to receive either 400 mg sorafenib or

placebo twice a day. Results from the study—the largest randomized controlled trial ever conducted

in advanced renal cell cancer—were presented in an oral session during the ASCO 2005 meeting in

Orlando. Disease progression was significantly delayed in those patients who received sorafenib. As

assessed by independent radiologic review, progression-free survival was doubled to a median value

of 24 weeks in patients receiving sorafenib as compared to 12 weeks for patients receiving placebo

(P-value < 0.000001).172 In July 2005, the companies completed the submission of a new drug

application (NDA) with the FDA for sorafenib for patients with RCC, or kidney cancer. Discussions

are also underway with regulatory agencies about registration in other territories. A phase III study

ANTI-ANGIOGENIC DRUGS * 495

has been recently initiated in patients with advanced liver cancer, and an additional phase III study

evaluating sorafenib in combination with carboplatin and paclitaxel to treat patients with advanced

metastatic melanoma will begin soon. In 2005, a phase II clinical trial sponsored by the NCI has

begun to evaluate how well sorafenib works alone and paired with gemcitabine and anti-metabolite,

currently the standard chemotherapy for pancreatic cancer.

Some efforts are being devoted to the development of inhibitors of bFGF receptors, although

they are still in preliminary stages of development. Among others, we could point out the drugs

SU4984, SU5402, and PD166866, that inhibit the FGFR kinase, and PD173074, which inhibits

FGFR and VEGFR2.89

The overexpression of EGF or its receptor plays a main role in essential processes for tumor

progression, such as cell proliferation, apoptosis metastasis, and resistance to anti-tumoral drugs. In

addition, some experimental evidence supports the role for EGF in angiogenesis, probably by

modulating VEGF production.173 For these reasons, there has been a considerable interest in the

search and development of drugs that are selective inhibitors of EGFR, which could be good

candidates for anti-tumoral drugs. The advanced stage of development of the humanized monoclonal

antibodies cetuximab (IMC-225, erbitux, ImClone Systems Incorporated, New York, NY,

www.imclone.com) should be emphasized. It binds competitively and with high affinity to EGFR,

competing for natural ligand-binding sites and causing receptor internalization and down-

regulation.174 Pre-clinical studies using human cancer xenografts growing either s.c. or

orthotopically in nude mice have demonstrated that cetuximab inhibits tumor growth, increasing

the survival of mice, and inhibiting tumor-induced angiogenesis.173 Clinical efficacy of IMC-C225

appears to involve multiple mechanisms, including inhibition of cell cycle progression, induction of

apoptosis, inhibition of angiogenesis, inhibition of metastasis, and enhancement of the response to

chemotherapy and radiotherapy.173 Very recently, it has been reported that cetuximab inhibits VEGF

secretion in cell culture and in animal models, and that this inhibition occurs at the level of

transcription in response to a reduced level of hypoxia inducing factor (HIF)-1a. These results justifyfurther testing of therapeutic strategies that combine cetuximab with approaches inhibiting the

function of VEGF or the VEGF receptor.175

Phase I studies of cetuximab both as a single agent or in combination with conventional

chemotherapy or radiotherapy confirmed a lack of immunogenicity of this compound, that is

generally well tolerated, with skin rashes and allergic reactions being the most clinically important

adverse events reported.173 Cetuximab has demonstrated in phase II/III studies that it can make the

cancer cells more sensitive to chemotherapy and radiation. Results of a large phase II study have

shown response when used in combination with irinotecan in 22.9% of patients with EGFR-

expressing, irinotecan-refractory CRC.176 Cetuximab was approved in February 2004 for this

indication by the US FDA, and for use in Switzerland, Iceland, Norway and the member states of the

European Union. Very recently, in a phase II study cetuximab in combination with carboplatin has

demonstrated clinical activity and an acceptable safety profile in heavily pre-treated patients with

recurrent or metastatic nasopharyngeal carcinomawho had previously experienced treatment failure

with platinum-based therapy.177 Other phase II and III studies showed significant response to

treatment invariable proportions of patients with RCC, carcinoma of the head and neck, NSCLC, and

pancreatic cancer, when cetuximab is used first or second line in combination with chemother-

apy.178–183 Results of an international, randomized phase III clinical trial of 424 patients who

received radiation � cetuximab for advanced head and neck cancer demonstrated a near doubling of

median survival for patients treated with radiation plus cetuximab, 54 months versus 28 months for

patients treatedwith radiation alone.184 Thus, cetuximab is emerging as avery promising new therapy

to be used in combination with existing therapies for the treatment of a spectrum of solid tumors.

Cetuximab is being currently evaluated in phase III in combination with standard chemotherapy in

CRC, and pancreatic cancer, in phase II/III in patients who have unresectable liver metastases from

colorectal cancer, and in phase II in NSCLC, CRC, ovarian and metastatic pancreatic cancer.185–189

496 * QUESADA, MUN‹ OZ-CHA¤ PULI, AND MEDINA

The encouraging data obtainedwith the EGFR inhibitor iressa (gefitinib,ZD1839, Astra Zeneca,

London, UK, www.astrazeneca.com) in phase II trials in advanced CRC, NSCLC, breast cancer,

glioblastoma, and head and neck cancer gave an impulse to the clinical development of this

compound, with more than 35 clinical studies under course in mid-2004.190–200 Iressa is being

evaluated in phase III inNSCLCand in squamous cell carcinoma of the head and neck, and in phase II

in ovarian, breast or skin cancer, metastatic bladder cancer, CRC, glioblastoma, progressive

metastatic neuroendocrine tumors, metastatic synovial sarcoma, recurrent or progressive CNS

tumors, and childhood brain stem glioma. Iressa has been used in over 200,000 patients since its first

launch in Japan in 2002 and is licensed in more than 30 other countries. In May 2003, it received the

US FDA accelerated approval for the third-line treatment of NSCLC. However, in December 2004,

AstraZeneca notified that a large clinical trial of nearly 1,700 patients comparing iressa with placebo

in patients with NSCLC who had failed other therapies failed to show a survival benefit

(www.fda.gov). The trial was looking for increased survival in patients, but despite statistically

significant improvement in tumor shrinkage, this did not translate into a significant survival benefit.

Recent data suggests that only a small subgroup of non-smoker patients with bronchioloalveolar

pathologic subtype showed a better response to iressa.201 The response rates significantly increased

among oriental patients, probably due to the existence of somatic mutations in the EGFR kinase

domain in those patients who responded well to iressa.202–204 Thus it is likely that ultimate approval

of iressa in lung cancer will only be for a small identifiable subset of patients with NSCLC.

The EGFR inhibitor erlotinib (OSI-774, tarceva, OSI Pharmaceuticals, Inc., Melville, NY,

www.osip.com), was Iressa’s closest competitor and is now in a very dominant market position.

Erlotinib received fast track status from the FDA in May 2002 for treatment of chemotherapy-naıve

advanced-stage NSCLC, which was later extended to include second- or third-line monotherapy in

patientswith incurableNSCLC failing standard chemotherapy. InNovember 2004, erlotinib received

US FDA approval for the treatment of NSCLC and gained the distinction of being the first EGFR

inhibitor to show a survival benefit in lung cancer patients. The FDA based its approval decision for

erlotinib on results from a randomized, double-blind, placebo-controlled pivotal phase III trial of

patients with second- and third-line advanced NSCLC. The trial included 731 patients with advanced

NSCLC for whom one or more chemotherapy regimens had failed. Patients receiving erlotinib

showed an increase in median survival by 42% compared to those in the placebo arm (6.7 months vs.

4.7 months), an improvement of 2 months.205 There was also a significant increase in both the length

of time before patients disease symptoms deteriorated and the time when patients were stable and

therewas no progression of their cancer. A 45% improvement in survival at 1 year was observedwith

erlotinib, with benefits being shown in a broad spectrum of patients. Similarly to the significant

survival benefit in NSCLC, erlotinib has also shown survival benefit in a phase III study in locally

advanced ormetastatic pancreatic cancer patients. In early 2005OSI Pharmaceuticals announced that

a multi-center, randomized, double-blind, placebo-controlled phase III trial evaluating erlotinib plus

gemcitabine chemotherapy in 569 patients with locally advanced ormetastatic pancreatic cancer met

its primary endpoint by demonstrating a statistically significant 23.5% improvement in overall

survival when compared to patients receiving gemcitabine plus placebo.206,207 Erlotinib is the first

drug shown in a phase III trial to prolong survival when added to the standard of care (gemcitabine) in

the treatment of patients with previously untreated advanced pancreatic cancer. Other indications in

which erlotinib has produced objective evidence of anti-tumor activity in patients failing standard

chemotherapy include ovarian cancer as well as cancers of the head and neck. Erlotinib is currently

being evaluated in an extensive clinical development program by a global alliance among OSI

Pharmaceuticals, Genentech, and Roche, and is being currently evaluated in more than 35 clinical

studies, including phase II in patients with NSCLC, glioblastoma multiforme, recurrent breast, head

and neck, ovarian, renal or colon cancer, glioblastomamultiforme, liver cancer, advanced esophageal

cancer or stomach cancer,metastaticmalignant peripheral nerve sheath tumor, and recurrent cervical

cancer.190,208–218

ANTI-ANGIOGENIC DRUGS * 497

A final commentary to the existence of drugs acting on sites of the signaling pathways that are

located downstream the angiogenic factor receptors, and that also exhibit anti-angiogenic properties.

Among others, we could mention LY317615 (Eli Lilly, Indianapolis, IN, www.lilly.com), which

inhibits the protein kinase C b, an enzyme that is involved in the signaling pathway of VEGFR2, and

that exhibits anti-angiogenic activities in the rat cornea assay and anti-tumoral activities in vivo in

glioblastoma and gastric, renal, colorectal, breast, ovary, and lung tumors.219–223 Phase I studies in

patients with solid tumors have been completed, and phase II studies in patients with glioma or

lymphoma are in progress.224

3 . A G E N T S T H A T I N H I B I T E N D O T H E L I A L C E L L P R O L I F E R A T I O N

The first experimental strategy for the screening of new anti-angiogenic compoundswas the search of

endothelial cell growth inhibitors. One of the firstly described inhibitors of angiogenesis, fumagillin

(Fig. 4) was isolated from an endothelial cell culture that had been contaminated by the fungus

Aspergillus fumigatus fresenius. Fumagillin inhibits endothelial cells proliferation in vitro, and

angiogenesis and tumor growth in experimental models. A prolonged administration of fumagillin is

limited by a severe weight loss. This fact aimed at the synthesis of new derivatives with improved

anti-angiogenic activities and lower undesired effects.225 The fumagillin analog AGM-1479/TNP-

470 (Fig. 4) (TAP Pharmaceuticals, Inc., Deerfield, IL) is more selective to endothelial cells than

fumagillin.226 AGM1470 mechanism of action seems to be related to the inhibition of type 2

methionine amino peptidase, and cyclin-dependent kinases.227,228 This drug is active in several

animal tumor models, and in a tumorogenesis model in transgenic mice, exhibiting therapeutic

Figure 4. Inhibitors ofendothelial cell proliferation.

498 * QUESADA, MUN‹ OZ-CHA¤ PULI, AND MEDINA

synergies with ionizing radiation in human glioblastoma multiforme xenographs.229,230 AGM1470

anti-tumor activity has brought it to phase II/III clinical studies with a series of solid tumors, and

phase I studies in acute leukemia, lymphoma, and Kaposi’s sarcoma. The appearance of secondary

toxicities (encephalopathy and ataxia) seems to be reversible after treatment interruption, and new

approaches to develop polymer conjugates may diminish drug-related toxicities.231 Partial responses

have been described in patients of renal carcinoma, cervix cancer, and Kaposi sarcoma.232–235 Some

partial responses have also been obtained after co-administration of AGM1470 and paclitaxel in the

treatment of lung cancer patients.236

Squalamine (MSI-1256F, Magainin Pharmaceuticals, Detroit, MI, www.magainin.com,

Genaera Corporation, PlymouthMeeting, PA, www.genaera.com) is an aminosterol (Fig. 4) isolated

from the liver of the shark Squalus acanthias. Squalamine inhibits angiogenesis and tumor growth in

multiple animal models. This effect is mediated, at least in part, by blocking mitogen-induced

proliferation and migration of endothelial cells, thus preventing neovascularization of the tumor.

Squalamine has no observable effect on unstimulated endothelial cells, is not directly cytotoxic to

tumor cells, does not alter mitogen production by tumor cells, and has no obvious effects on the

growth of newborn vertebrates. Squalamine was also found to have remarkable effects on the

primitive vascular bed of the CAM.237 Squalamine is an inhibitor of Naþ/Hþ interchanger protein.238

Evenwhen no limiting toxicities have been found in phase I studies, no objective responses have been

reported for squalamine as a single agent. Some positive results have been obtained in phase II studies

in lung cancer in combination with standard chemotherapy. Strong clinical results, including

improved vision for the phase I/II trial have been reported for the treatment of aged-related macular

degeneration. Squalamine is currently being evaluated in phase II studies in solid tumors (NSCLC

and prostate) co-administered with chemotherapy in ovarian cancer patients. Its big potential as a

non-invasive anti-angiogenic therapy for blinding eye disease, is being evaluated in aged-related

macular degeneration, being granted in October 2004with the Fast Track designation by FDA for the

treatment of this disease.239,240

Rapamycin (sirolimus, Fig. 4), a well-known immunosuppressor, inhibits tumor growth and

angiogenesis in in vivo models. This anti-angiogenic activity has been related to the inhibition of

endothelial cell proliferation after VEGF stimulation, and with a decrease in the production of this

angiogenic factor.241 Rapamycin is an inhibitor or the mammalian target of rapamycin (mTOR), a

downstream effector of the phosphatidylinositol 3-kinase (PI3K)/Akt (protein kinase B) signaling

pathway,whichmediates cell survival and proliferation.As a result of its positionwithin this pathway

and its central role in controlling cellular growth, mTOR is viewed as an important target for anti-

cancer therapeutics development.242 Everolimus (RAD-001, SDZ RAD, Certican; Fig. 4) (Novartis

Pharma, Basel, Switzerland, www.novartis.com), is an analog of rapamycin that can be administered

orally, and exhibits more favorable pharmacokinetic properties.243 This compound is very selective

to endothelial cells, with a significantly lower activity on non-endothelial cell lines.244 Everolimus

presents a limited toxicity in patients. It is being developed as an anti-proliferative agent and has been

approved in Europe as an immunosuppressive agent in the solid organ transplantation setting.242–246

In the last few years, several tumor-produced endogenous molecules with angiostatic activity

have been described. Their discoverywas based on the observation that some primary tumors seemed

to inhibit their own metastases; the primary tumor removal provoked the growth of the metastases.

Recent evidence suggests that physiological levels of endogenous inhibitors of angiogenesis can

serve as endothelium-specific tumor suppressors.247 The search of circulating factors produced by

tumors brought to the isolation of angiostatin and endostatin (EntreMed, Rockville, MD,

www.entremed.com).248,249 Angiostatin was isolated from the urine of lung cancer bearing mice.

Those mice only suffered from metastasis when the primary tumor was removed. Angiostatin has

been suggested to be produced by partial hydrolysis of plasminogen, probably by means of a

macrophage metalloelastase or other proteases produced by tumor cells.250,251 Endostatin was

initially found in the medium conditioned by hemangioendothelioma cells, and it can be originated

ANTI-ANGIOGENIC DRUGS * 499

by proteolysis of collagen XVIII, frequently present in the basal membranes. A variety of murine

tumors as well as human breast, prostate, and colon tumors in human xenograft models regressed

when treatedwith angiostatin or endostatin. Regression of tumors upon systemic treatmentwith these

proteins was in part due to increased tumor cell apoptosis. Repeated cycles of endostatin therapy led

to prolonged tumor dormancy without further treatment and were not associated with any apparent

toxicity or acquired drug resistance.252 Endostatin-mediated apoptosis is associated with several

apoptotic signaling pathways including overloading of intracellular magnesium and calcium, as well

as regulation of p53 and Bcl-2 expression.253 Endostatin was the first angiogenesis inhibitor in

reaching the clinical studies. Phase I clinical studies indicated that endostatin is well tolerated by

patients, but the results were not as satisfactory as expected, since evenwhen some evidence ofminor

anti-tumor activity was observed, no objective response was obtained in those studies.254–257 New

hopes are arising fromvery recently reported phase III results showing that the addition of endostar (a

new recombinant human endostatin developed by Medgenn Co., Yantal, Shandong, China,

www.medgenn.com) to vinorelbine and cisplatin (NP) regimen resulted in significant and clinically

meaningful improvement in response rate, median TTP, and clinical benefit rate compared with NP

alone in advanced NSCLC patients.258 Angiostatin has been well tolerated in phase I clinical studies,

and is being evaluated in phase II for the treatment of lung cancer patients in combination with

paclitaxel and carboplatin.259,260 A gene-based angiostatin treatment for vision loss is also being

explored.261 Efforts are being devoted to the development of alternative ways of administration to

increase angiostatin or endostatin efficacy. They include the use of osmotic pumps for the continuous

administration of endostatin, and intravenous administration of angiostatin and endostatin genes

complexed to cationic liposomes.262–264

4 . A G E N T S T H A T I N H I B I T E N D O T H E L I A L C E L L I N V A S I O NA N D M O T I L I T Y : P R O T E A S E I N H I B I T O R S

Drugs that inhibit matrix metalloproteinases (MMPs) have been recently compared to a mirage, a

tantalizing possibility that keeps always out of reach. In theory,MMPs are one of the most promising

targets to fight cancer and other diseases, but the clinical failures of their inhibitors do not support this

idea.265,266 The discovery of the relation between MMPs overexpression and tumor invasion awoke

the interest in the search of inhibitors ofMMPs that, in theory, could act on at least two steps of tumor

progression: angiogenesis and metastasis. But time has demonstrated that things are not so simple:

MMPs are not only involved in the extracellular matrix, basement membrane, and connective tissue

remodeling, but they play a relevant role in the control of other molecules that regulate cell

proliferation, migration and differentiation, angiogenesis and apoptosis. This may make the

prediction of the consequences derived from the inhibition of a given protease difficult, since normal

cell processes can be affected.267

This complex panorama was not known when the first MMPs inhibitors were described. They

were designed to contain a hydroxamate group that would complex the metalloprotease Zn2þ,

essential for its activity. Batimastat (Fig. 5) (BB94, British Biotech, Oxford, UK, www.britishbio-

tech.com), awide spectrum, competitive and reversible inhibitor ofMMPs, was the first compound of

this family to reach clinical studies.268 Its development was interrupted in 1996 to favor that of

marimastat (Fig. 5) (BB2626,BritishBiotech, licensed to Schering-PloughCorporation,Kenilworth,

NJ, www.schering-plough.com, and Tanabe Seiyaku Co, Osaka, Japan, www.tanabe.co.jp), a

compound that maintained the MMP inhibitory properties of batimastat, but with an increased

solubility that could allow an oral administration. After promising results in pre-clinical and early

clinical studies, undesired effects including fatigue and cumulative poliarthritis forced to reduce the

doses in subsequent studies. Unfortunately, the newdoseswere not high enough to reach a therapeutic

efficacy, and results obtained in phase III clinical studies were very disappointing (marimastat failed

500 * QUESADA, MUN‹ OZ-CHA¤ PULI, AND MEDINA

to show superior efficacy over either standard chemotherapy or placebo in pivotal phase III trials in

glioblastoma, breast, ovarian, and small and non-small cell lung cancer).269,270 In two recent phase III

studies with SCLC and metastatic breast cancer patients, marimastat treatment did not only fail in

improving survival, but had a negative impact on quality of life.271,272

A similar hapless fate was that of the drug prinomastat (AG3340, Agouron Pharmaceuticals,

Inc., San Diego, CA, www.agouron.com), another synthetic derivative of hydroxamic acid (Fig. 5)

that exhibits a more selective MMPs inhibition profile. Pre-clinical studies showed that prinomastat

produced growth delays in a variety of tumors including neuroblastoma, as well as in cancers of the

lung, breast, colon, brain, and prostate. Prinomastat inhibited tumor angiogenesis and cell

proliferation in neuroblastoma and enhanced the efficacy of carboplatin and taxol in NSCLC.273,274

In spite of the fact that the compound seemed to be well tolerated and synergize with the traditional

chemotherapy in early clinical trials, the most frequent side effects being musculoskeletal pain and

stiffness, prinomastat development was almost interrupted in August 2000, after discouraging results

in phase III clinical studies for the treatment of prostate and lung cancer patients.275–280 Data from

over 1,000 patients with stage IIIB, IV, or recurrent NSCLC receiving gemcitabine/cisplatin or

paclitaxel/carboplatin showed that prinomastat doubled the risk of venous thromboembolisms in

patients with advanced NSCLC receiving chemotherapy.281 New hopes for the clinical usefulness of

this compound may derive from its use in combination with photodynamic therapy or in

ophthalmology and dermatology.282–286

The drug BAY12-9566 (Fig. 5) (Tanomastat, Bayer, Inc., Leverkusen, Germany, www.bayer.-

com) is structurally different to the above-mentionedMMP inhibitors. It is a butanoic acid derivative

that selectively inhibits MMP2, MMP3, and MMP9, which could explain its lower muscular

toxicity.287–289 Promising pre-clinical data included inhibition of tumor growth and metastasis in

murine melanoma and human colon tumor xenografts, and phase I clinical studies indicated that the

compound was relatively well tolerated.290 Even when the compound reached phase III clinical

Figure 5. Inhibitors ofmatrixmetalloproteinases.

ANTI-ANGIOGENIC DRUGS * 501

studies for the treatment of lung, pancreas and ovary cancers, a decrease in the survival of the patients

treated with this compound provoked an immediate interruption of its clinical development.291

Nowadays the pharmaceutical companies do not slack in their effort for finding a new inhibitor of

MMPs with a wide clinical application. British Biotech is developing new inhibitors, such as

solimastat (BB-3644, Fig. 5), a wide spectrum inhibitor, orally administrable and with a series of

advantages when compared tomarimastat, that is, lower effects on skeletal muscle, and an additional

inhibition of the production of TNFa. BB-3644 has been evaluated phase I clinical studies for the

treatment of solid tumors that are refractory to chemotherapy. As dose-limiting musculoskeletal

toxicity was encountered at doses of BB-3644 unlikely to provide an advantage over currently

available MMPIs, further evaluation is not recommended.292,293

BMS-275291 (Fig. 5) (Bristol Myers Squibb, New York, NY, www.bristolmyers.com) is a

peptidomimetic soluble MMPI that contains a chemically novel mercaptoacyl zinc-binding group

and that was rationally designed to inhibitMMPs activities, but not those of other proteases that could

be involved in the release ofmolecules including TNFa, L-selectine, and the receptors of TNFa or IL-6. In vitro, BMS-275291 is a strong inhibitor ofMMP1,MMP2,MMP7,MMP9, andMMP14. BMS-

275291 inhibits tumor growth and metastasis in murine models, and inhibits angiogenesis in the

Matrigel plug angiogenesis model.294 BMS-275291 does not inhibit the sheddases, which are related

metalloproteinases that regulate pro-inflammatory cytokine and cytokine receptor shedding from the

cell surface.295 Shedding of tumor necrosis factor and its receptor (TNF-RII) were hypothesized to

play a role in MMPI-associated musculoskeletal toxicity. In early clinical trials, BMS-275291

therapywas not associatedwith arthralgia ormyalgia.296–297Recent results from a randomized phase

II trial in 80 hormone refractory prostate cancer patients with bone metastasis indicated that BMS-

275291 was well tolerated and had no dose-limiting arthritis.298 Phase II/III studies with BMS-

275291 in combination with paclitaxel and carboplatin for the treatment of advanced NSCLC

patients and in phase II in combination with zoledronate in patients with hormone refractory prostate

cancer are under curse. BMS-275291 plus paclitaxel/carboplatin was well tolerated and active in

advanced non-small cell lung cancer. Treatment with BMS-275291 was not limited by drug-related

arthrotoxicity, and tumor response was as expected. Patient accrual was continued to further

investigate the effect of BMS-275291 on overall and progression-free survival in a phase III

setting.299,300 Very recently reported results from this randomized phase III study of BMS-275291 in

combination with paclitaxel and carboplatin in 774 patients of advanced NSCLC, indicated that

BMS-275291 added to chemotherapy increased toxicity and did not improve survival in those

patients.301 A randomized phase II feasibility trial was performed to evaluate the safety, phar-

macokinetics and feasibility of incorporating BMS-275291 into adjuvant breast cancer therapy. The

pattern of arthralgia in BMS-275291-treated patients was consistent with matrix metalloproteinase

inhibitor toxicity, showing that an adjuvant trial in this patient population is not feasible. Although the

rate of arthralgia ormyalgiawas sufficient to require early termination of the trial, it was nevertheless

lower than reported for other MMPIs. Therefore, although the rational design of this MMPI to avoid

sheddase release did not eliminate arthralgia, it may have widened the therapeutic window. The

critical question, whether it is possible to completely separate inhibition of MMPs that are important

in cancer progression from those whose inhibition produces joint toxicity, still remains.302

MMI270 (Fig. 5) (CGS27023A, Novartis Pharma AG, Basel, Switzerland) is a potent, broad

spectrum, orally active MMPI that inhibits the gelatinases MMP2 and MMP9 and the stromelysin

MMP3 in low nanomolar concentrations. In rat tumor models of breast and endometrial cancer,

MMI270 as a single agent or in combination with megestrol acetate significantly reduced tumor

burden and number of lymph node and lung metastases.303 MMI270 inhibited the migration and

invasion of endothelial cells, in addition to tube formation in vitro.304 A single-agent phase I study of

MMI270 performed in patients with advanced solid cancer indicated that it was rapidly absorbed

following oral administration, reaching in plasma of patients those concentrations required for

optimum MMPI activity. The pattern of toxicity (arthralgia and/or myalgia) was similar to that

502 * QUESADA, MUN‹ OZ-CHA¤ PULI, AND MEDINA

described with other broad-spectrumMMPIs.305 Considering that althoughMMPIs may be active as

single agents, it is likely that they will bemore effective in combination with chemotherapy. In fact, a

dose-finding study of MMI270 combined with 5-FU and folinic acid has already been conducted.

Results from this study indicate that MMI270 can be safely administered in conjunction with 5-FU

and folinic acid over a range of dose levels.306 However, no clear responses have been found in the

mentioned clinical trials.

The first positive results of clinical trials withMMPs inhibitors were reported in 2001. In a study

with 414 non-operable pancreas cancer patients, marimastat treatment resulted to be as effective as

the treatment with gemcitabine, the reference compound for this type of tumors.270 Nevertheless in

subsequent studies the therapeutic benefit of the use of marimastat in combination with gemcitabine

versus gemcitabine alone has not been established.307 The most hopeful results are derived from a

study with 367 patients of gastric cancer, where it could be demonstrated that the treatment with

marimastat increased patient survival. This therapeutic benefit was more pronounced in those

patients that had previously been treatedwith chemotherapy.308More recently, newhopes have arisen

after partial responseswere obtainedwith the combination ofmarimastat and standard chemotherapy

in the treatment of patients of glioblastoma or pancreas and gastric cancers.309,310

The drug COL-3 (Fig. 5) (Metastat, CollaGenex Pharmaceuticals, Inc., Newtown, PA, www.

collagenex.com), is a modified tetracyclin which is devoid of anti-microbial properties and is a

competitive and selective inhibitor ofMMP2 andMMP9 enzymes. COL-3 also inhibits the activity of

activated neutrophil gelatinase, and blocks the invasion of several types of tumors into Matrigel and

basementmembranematrices. In addition, impressivegrowth-inhibitory activity has been noted after

COL-3 treatment in a broad range of human tumor cell lines and xenografts, including human

melanoma and metastatic prostate cancer, probably due to the induction of apoptosis.311–315 The

main undesired effect of this compound is the photosensibilization induction, so that it has to be

administered in combination with the use of sun protection filters.316 Although no objective anti-

tumor responses were observed, apparent clinical benefit in several patients with soft tissue sarcoma

and with advanced solid malignancies that were unresponsive to standard therapy, as well as a 44%

overall response rate in patients with AIDS-related Kaposi’s sarcoma, suggest that COL-3 should

undergo additional evaluation for the treatment of various types of sarcoma and othermalignancies in

whichMMPs may play an important role in invasion, proliferation, and metastases.316–318 This drug

is being tested in phase I/II studies with astrocitoma/multiforme glioblastoma, and in phase II with

Kaposi sarcoma patients, in collaboration with the NCI (www.clinicaltrials.gov).

Neovastat (AE-941, Aeterna Laboratories Inc., Quebec, Canada, www.aeterna.com) is a shark

cartilage extract soluble in aqueous media, representing less than 5% crude cartilage. It is a multi-

functional anti-angiogenic product (containing several molecules with biological activity), which

affects multiple levels of the angiogenic cascade. Neovastat inhibits MMP2, MMP9, and MMP12,

interferes with VEGFR2 signaling pathway, stimulates tissue-type plasminogen activator enzymatic

activity, inhibits endothelial cell proliferation and tubulogenesis, and induces apoptosis in

endothelial cells.319,320 This pro-apoptotic activity is specific for endothelial cells and correlates

to an increase in caspases 3 and 8.321 Anti-angiogenic activity of the compound is also supported by

its inhibitory effect on neovascularization in the ex ovo CAM model as well as by its inhibition of

neovascularization induced in vivo by s.c. implants of Matrigel.322 Chronic administration of

Neovastat significantly inhibits tumor growth in vivo andmetastases invasion in an in vivoLewis lung

carcinoma model.314 Neovastat is active in the treatment of multiple myeloma, renal and lung

carcinoma, with an increase in the patients survival time, without the appearance of great

toxicities.323 A significant survival advantage has been observed in NSCLC patients receiving the

highest doses of neovastat in a phase I/II trial.324 The main activity has been on neovastat in clinical

trials for renal and NSCLC, both reaching phase III.325–327 Results from a phase III clinical trial

evaluating neovastat in 305 patients with advanced renal cell cancer refractory to immunotherapy did

not meet its primary endpoint of improving median survival time. Despite the drug’s failure,

ANTI-ANGIOGENIC DRUGS * 503

significant survival advantage was observed in a subgroup of certain healthier patients. This

38-patient subgroup showed a median survival time of 26.3 months for those treated with Neovastat,

comparedwith 12.6months for patients receiving a placebo. The increase inmedian survival time for

the renal cell carcinoma subgroup, which had an earlier disease stage compared with the overall

group, seems to suggest that treatment with neovastat in earlier-stage cancer could be more

efficacious.328 Aeterna has decided to focus neovastat efforts in lung cancer only. A phase III trial,

sponsored by theNCI and scheduled to be finished in 2006, is evaluating neovastat inNSCLCpatients

in combination with other drugs and radiotherapy.329 Neovastat therapeutic potential is not restricted

to cancer, since good results have been obtained with this drug in phase II studies for the treatment of

psoriasis patients.330

Finally, a brief commentary about a new experimental approach to inhibit the proteolytic

activity, consisting of the use of drugs that may inhibit the expression, secretion or activation of

proteases, or increase those of their inhibitors. Among them, we could mention curcumin, a

polyphenol from the plant Curcuma longa of widespread use as spice or alimentary colorant,331,332

halofuginone, an alkaloid from the plant Dichroa febrifuga, that inhibits MMP2 expression by

interfering TGF-b signaling pathway,333 and aeroplysinin-1, isolated from the marine sponge

Aplysina aerophoba, that induces a shift in the proteolytic balance toward anti-proteolysis, and

therefore to less invasive phenotypes.334

5 . A G E N T S T H A T I N H I B I T E N D O T H E L I A L C E L L A D H E S I O N

Another strategy for the anti-angiogenic therapy consists of the inhibition of endothelial cell adhesive

interactions during their migration. Endothelial cell migration depends on adhesion to the

extracellularmatrix proteins bymeans of a number of cell adhesion receptors known as integrins. The

avb3 integrin, a receptor for extracellular matrix components that contains the peptidic motif

arginine-glycine-aspartic acid (RGD), is an attractive target for the anti-angiogenic therapy, since it is

only present in the cell surface of the activated endothelial cells, and not in quiescent endothelia and

other cell types. It was initially described that some peptides that are antagonist of RGD domain, and

monoclonal antibodies generated against the avb3 integrin (LM609) disrupted ongoing angiogenesis

on theCAM, leading to the rapid regression of histologically distinct human tumors transplanted onto

the CAM.335 Intravenous administration of LM609 in a SCID mouse/human chimerical model with

transplanted human skin containing avb3-negative human breast tumor cells, either prevented tumor

growth ormarkedly reduced tumor cell proliferationwithin themicroenvironment of the human skin.

These LM609-treated tumors not only contained significantly fewer human blood vessels but also

appeared considerably less invasive than tumors in control animals.336 The excellent results obtained

in animal models led to the evaluation of integrin antagonists in clinical trials. Those conducted with

vitaxin (MEDI-522, Scripps Research Institute, la Jolla, CA;MedImmune, Gaithersburg,MD; Ixsys,

La Jolla, CA) are remarkable. Vitaxin is a humanized form of the LM609 antibody that selectively

binds to sprouting blood vessels and suppresses tumor growth in several animal models.336,337 Based

on the selectivity of avb3 for the activated endothelial cells and on their anti-angiogenic activity in

vivo, some clinical assays have been carried out to establish vitaxin tolerability and pharmacokinetic.

Vitaxin antibodies have been well tolerated by cancer patients, giving positive results in some

cases.92,338Very recently, results from amulti-center, non-comparative phase II study have suggested

a potential clinical activity of vitaxin with or without dacarbazine in patients with metastatic

melanoma.339 Phase II studies for the treatment of leiomyosarcoma patients, and other clinical

studies for the treatment of inflammatory diseases and rheumatoid arthritis are in progress.340–342

A different way to block the avb3 receptor is by means of RGD-type peptides. Their use may

present some advantages when compared to that of antibodies, such as their easy synthesis, resistance

to proteolysis, and low immugenicity. Among others, we could mention the cyclic peptides

504 * QUESADA, MUN‹ OZ-CHA¤ PULI, AND MEDINA

EMD12974 (cilengitide, RGDf-[NmeVal) and EMD270179 (RGDf-ACHA) (Merck KGaA,

Darmstadt, Germany, www.merck.de). EMD12974 has demonstrated efficacy in melanoma tumor

models as a single agent,343 and its combination with radio immunotherapy significantly increased

efficacy and tumor and endothelial cell apoptosis without apparent increases in toxicity.344

EMD12974 has reached phase II in NSCLC, pancreatic cancer, and glioblastoma.345–349 In January

2004, orphan designation was granted by the European Commission toMerckKGaA for EMD12974

for the treatment of glioma. This drug is currently being evaluated in phase I in patientswith advanced

tumors and lymphoma, and in children with refractory primary brain tumors and in phase II in

patients with metastatic prostate cancer and recurrent glioblastoma multiforme.

Some efforts are being invested in the search of non-peptidic, small size molecules that are

antagonist ofavb3. Severalof thesedrugshavedemonstrated agoodanti-angiogenic activity in invitro

and in vivo models, but they have not yet reached the clinical phases of development.350–354

Finally, we will make a reference to the recent controversy concerning the hypothesis that avb3and/or avb5 integrins are necessary for tumor angiogenesis, since their absence in knockout mice

causes increased angiogenesis and tumor growth. This apparent paradox suggests that integrins are

also negative regulators of angiogenesis and therefore, anti-integrin drugs may act mainly as agonist

of somenegative regulator functions. Thus, although integrinsmay still be considered as a valid target

for the pharmacological inhibition of angiogenesis, the design and development of the new anti-

integrin drugs will have to take into account all the molecular processes that could be affected by

them.355

6 . O T H E R A G E N T S T H A T I N H I B I T A N G I O G E N E S I S

A. Thalidomide

Thalidomide (a-(N-ftalimido)glutarimide, Fig. 6) (EntreMed, Ocknille, MD Celgene Corporation,

Warren, NJ, www.celgene.com) is an inhibitor of the bFGF- and VEGF-induced angiogenesis. It is a

chiral compound and the S(�) enantiomer presents strong teratogenic and anti-angiogenic

properties. It was initially used as sedative and anti-emetic in pregnant women, but it was given

up in 1961 because it provoked fetal malformations.356 However in 1965 its evaluation for the

treatment of inflammatory and autoimmune diseases was activated.357 The first reference of the anti-

angiogenic properties of thalidomide is due to D’Amato and colleagues, which found that this

compound inhibited the bFGF-induced angiogenesis in rabbit cornea.358 Even when thalidomide

mechanismof action is not clear, itmay be partially related to its capability to inhibit the expression of

some adhesion molecules, including VCAM-1 (CD106) and E-selectin, and mediated by ceramide

through depletion of VEGFRs, and antagonized by sphingosine-1-phosphate.359,360 Several clinical

studies have been conducted to evaluate the interest of thalidomide for the treatment of hematological

or solid tumors. Some positive results have been obtained with multiple myeloma patients, and

partially positive results have been obtained in the treatment of high grade glioma patients.361–364

Some hopeful results have been obtained in the treatment of Kaposi’s sarcoma, prostate cancer,

myelofibrosis withmyeloidmetaplasia, and neurofibromatosis.365–371 In a phase II study designed to

evaluate the anti-tumor activity and toxicity of thalidomide as a single agent in patients with

metastatic melanoma this compound showed poor activity, but acceptable toxicity.372 In spite of

some evidence pointing to the activity of thalidomide in renal carcinoma, the high toxicity of the

compound at doses needed for the anti-angiogenic activity makes its use unadvisable for the

treatment of this type of tumors.373–375 No positive results have been obtained in the treatment of

metastatic breast cancer patients.376 Recent results from a phase III study show that thalidomide does

not improve survival in patients receiving cranial irradiation (WBRT) for multiple brain

metastases.377 There are currently more than 50 clinical studies under curse with thalidomide. It is

being evaluated in combination with standard therapies in phase III in prostate, ovarian and renal

ANTI-ANGIOGENIC DRUGS * 505

cancer, SCLC, and multiple myeloma, and in phase II in multiple myeloma, hematological cancer,

glioma, glioblastoma, liver cancer, myelodysplastic syndrome, uterine sarcoma, and SCLC.

B. Carboxyamido-Triazole (CAI)

Carboxyamido-triazole (CAI) (Fig. 6) is a lowmolecular weight synthetic compound that inhibits the

cellular calcium input. It inhibits both endothelial cell proliferation and invasion, through inhibition

of endothelial adhesion and spreading, inhibition ofmigration and induction of apoptosis.378–381CAI

decreases the expression of MMP2 and VEGF in vitro, and that of VEGF and IL-8 in animal

models.382,383 CAI inhibits vascular tube formation in Matrigel and aortic ring assays and

angiogenesis in the chicken chorioallantoic membrane assay by blocking the calcium-dependent

nitric oxide synthase VEGF pathway.380–383 In clinical trials, CAI stabilizes and in some cases

reduces the tumoral mass and the metastatic spread at plasma concentrations that are in the range of

those that regulate the cell signaling in vitro.384,385 CAI anti-angiogenic and anti-metastatic

properties, together with its possible oral administration has led it to be tested in clinical trials, with

moderate positive results.385–388 CAI is currently being evaluated in combination with paclitaxel in

phase I in patients with advanced solid tumors or refractory lymphomas.

C. NM-3

NM-3 (Fig. 6) (ILEXOncology, Boston,MA,www.ilexonc.com) is an isocumarinic derivative orally

administrable that inhibits hypoxia-induced VEGF expression in human fibroblasts and a number of

tumor cell lines.389 NM-3 inhibits the proliferation of human umbilical vein endothelial cells in vitro,

Figure 6. Othermiscellaneous inhibitors ofangiogenesis.

506 * QUESADA, MUN‹ OZ-CHA¤ PULI, AND MEDINA

at concentrations 10-fold less than those required to inhibit normal fibroblasts or tumor cells (HT29,

MKN28, andMCF-7), and this effect is mediated by the generation of reactive oxygen species.390,391

NM-3 inhibits endothelial sprouting and tube formation in vitro, and exhibits in vivo activity against

Lewis lung carcinoma and human tumor xenografts, potentiating the anti-tumor effects of

chemotherapy and radiotherapy without an increase in toxicity.391–393 NM-3 is currently being

evaluated in phase I in patients with advanced solid tumors.

D. Tecogalan

Tecogalan (DS-4152) (Daiichi Pharmaceutical Corp., Fort Lee, NJ) is a polysaccharid–

peptidoglycan complex, isolated from the culture medium of Arthrobacter bacteria. Tecogalan

inhibits the bFGF-stimulated proliferation, migration, and tube formation of bovine choroidal

endothelial cells in vitro.394 It inhibits angiogenesis in the CAM, the corneal neovascularization

induced by bFGF, and the murine tumor M5076 cell-induced s.c. angiogenesis.395 Tecogalan

significantly inhibited the growth of s.c.-implantedB16melanoma, and inhibited the growth of tumor

xenografts of human breast cancer cells.395,396 Tecogalan anti-angiogenic activity has been proposed

to be related to its ability to block the angiogenic factors that bind heparin (bFGF, among others) as

well as the inhibition of proteases (u-PA and MMP1).397 After being assayed in phase I in patients

bearing tumors that are refractory to chemotherapy, it has been discontinued.398

E. Aeroplysinin-1

Aeroplysinin-1 is a bromotyrosine that can be isolated from several marine sponges. It inhibits

angiogenesis in vitro and in vivo, mainly due to the inhibition of endothelial cell differentiation.

Aeroplysinin-1 induces apoptosis in proliferating endothelial cells, and modifies their proteolytic

balance toward less invasive phenotypes, which results in an inhibition of the endothelial cells

migration and invasion in vitro.334 Some studies devoted to the evaluation of the aeroplysinin anti-

tumoral and anti-angiogenic activities in vivo, as well as the determination of its possible mechanism

of action, are under course.

F. Use of ‘‘Traditional’’ Chemotherapeutic Agents as Inhibitors of Angiogenesis

The increasing interest in the search of new strategies for inhibiting angiogenesis has provoked the

evaluation for anti-angiogenesis of many cytotoxic compounds traditionally used in cancer

chemotherapy. In spite of many chemotherapeutic drugs that have been reported to exhibit anti-

angiogenic properties, the clinical relevance of these results is unclear.

Miller et al. have proposed some criteria to consider a given drug useful for anti-angiogenic

therapies: (i) the compound is toxic for angiogenic endothelial cells at doses lower than those

required for killing tumor cells, (ii) it interferes with any activated endothelial cell functions without

causing cell death, (iii) it interferes specifically with any of the angiogenesis steps, and (iv) it inhibits

angiogenesis in in vivo assays.399

Among the number of anti-tumoral compounds being evaluated for anti-angiogenic therapies,

the taxanes stand out. Two taxanes are currently being studied in the clinic.400–403 Paclitaxel (taxol)

and docetaxel (taxotere), Figure 6, have demonstrated anti-tumoral activity in several solid

tumors,403–405 acting as potent radiosensibilizators.406–409 It has been recently reported that the

endothelial cells are 10–100 times more sensitive than tumor cells to these compounds. In addition,

these compounds mainly inhibit the proliferation and differentiation of endothelial cells, inducing

apoptosis, with docetaxel being ten times more active than paclitaxel. All these data should be taken

into accountwhen designing new clinical trials with taxanes as angiogenesis inhibitors.410 Therefore,

while traditional chemotherapy is normally administrated at the maximum tolerated doses, followed

by long resting periods, the chemotherapy ‘‘anti-angiogenic administration’’ advises the use of

prolonged expositions to low concentrations of the drug.411

ANTI-ANGIOGENIC DRUGS * 507

7 . C O N C L U D I N G R E M A R K S : T H E F A L L A N D R I S E O FA N T I - A N G I O G E N I C D R U G S

Angiogenesis inhibition has been proposed as a general strategy to fight cancer and other

angiogenesis-dependent diseases.412 However, in spite of the promising pre-clinical results, a first

generation of anti-angiogenic compounds gave poor results in clinical trials.413 Conceptual errors

and mistakes in the design of trials and in the definition of clinical endpoints could account for these

negative results.414 The discovery and pharmacological development of future generations of

angiogenesis inhibitors will benefit from further advances in the understanding of the mechanisms

involved in human angiogenesis. New styles of trials are necessary, to avoid missing potential

therapeutic effects. Different clinical endpoints, new surrogate biomarkers, and methods of imaging

will be helpful in this process.413–416 As shown in the case of the first relevant successful phase III

trial involving an anti-angiogenic compound,10 real efficacy in clinical trials may come with the

combined use of anti-angiogenic agents with conventional chemotherapy or radiotherapy, and

combinations of several anti-angiogenic compounds with different mechanisms of action.415

Several strategies to increase efficacy of anti-angiogenic therapy may be followed. They include

metronomic chemotherapy (continuous low-dose chemotherapy), and combination of anti-

angiogenic drugs and anti-HIF drugs, among others. Pre-clinical results indicating that anti-

angiogenic therapies are more effective in earlier than in later stages of the disease, could help to

understand the failure of so many clinical trials with patients in late-stage disease, and advised

new trials to be carried out with patients in earlier stages. However,more recently it has been reported

that potent VEGF blockade can cause regression of established tumors and metastases, indicating

that new generations of anti-VEGF drugs may be effective as single agents in the treatment of bulky

advanced-stage cancer.

On the other hand, one of the main concerns regarding the problems that could derive from an

anti-angiogenic therapy is now being discarded. By targeting endothelial cells, the anti-angiogenic

therapy could cause hypoxia therefore provoking an activation of cancer cells, and increasing their

capability to induce angiogenesis, to invade and metastasize. Recent results demonstrate that

although a shift from normoxia to hypoxia indeed activates cancer cells for aggressive behavior, most

cancer cells are constitutively activated by physiological hypoxia, and anti-angiogenic therapy may

not galvanize them further.417

As originally conceived, anti-angiogenic therapy would inhibit the formation of new blood

vessels without affecting the pre-existing ones. However, recent evidences indicate that some

angiogenesis inhibitors do not only block angiogenesis in tumors, but can also change or destroy

abnormal tumor vessels, causing vessel regression and normalization as well as stromal

maturation resulting in a reversion to a non-invasive tumor phenotype.418,419 The vascular

normalization process prunes immature vessels and improves the integrity and function of the

remaining vasculature by enhancing the perivascular cell and basement membrane coverage, and

induces a hydrostatic pressure gradient across the vascular wall, which leads to a deeper

penetration of molecules into tumor.420 In this way, certain anti-angiogenic agents could

transiently ‘‘normalize’’ the abnormal structure and function of tumor vasculature to make it more

efficient for oxygen and drug delivery.421 This mechanism might contribute to the frequently

observed potentiation of conventional therapies by anti-angiogenic agents. The normalization

process is mediated by angiopoietin-1 and matrix metalloproteinases and creates a window of

opportunity for improved sensitivity to ionizing radiation and the delivery of chemotherapeutic

drugs.422

Finally, the existing anti-angiogenic strategies should include other alternative and

complementary approaches, such as vascular targeting and angioprevention.423,424 The increasing

number of new anti-angiogenic drugs entering clinical trials clearly shows that the initial

disappointment has been overcome. Exciting new results are awaited in the near future.

508 * QUESADA, MUN‹ OZ-CHA¤ PULI, AND MEDINA

R E F E R E N C E S

1. Gasparini G. The rationale and future potential of angiogenesis inhibitors in neoplasia. Drugs 1999;58:17–38.

2. Kerbel RS. A cancer therapy resistant to resistance. Nature 1997;390:335–336.3. Brekken RA, Overholser JP, Stastny VA, Waltenberger J, Minna JD, Thorpe PE. Selective inhibition of

vascular endothelial growth factor (VEGF) receptor 2 (KDR/Flk-1) activity by a monoclonal anti-VEGFantibody blocks tumor growth in mice. Cancer Res 2000;60:5117–5124.

4. Gerber HP, Ferrara N. Pharmacology and pharmacodynamics of bevacizumab as monotherapy or incombination with cytotoxic therapy in preclinical studies. Cancer Res 2005;65:671–680.

5. Kim KJ, Li B, Winer J, Armanini M, Gillett N, Phillips HS, Ferrara N. Inhibition of vascular endothelialgrowth factor-induced angiogenesis suppresses tumour growth in vivo. Nature 1993;362:841–844.

6. Fernando NH, Hurwitz HI. Targeted therapy of colorectal cancer: Clinical experience with bevacizumab.Oncologist 2004;9(Suppl 1):11–18.

7. Burke PA,DeNardo SJ.Antiangiogenic agents and their promising potential in combined therapy. Crit RevOncol Hematol 2001;39:155–171.

8. Kabbinavar F, Hurwitz HI, Fehrenbacher L, Meropol NJ, Novotny WF, Lieberman G, Griffing S,Bergsland E. Phase II, randomized trial comparing bevacizumab plus fluorouracil (FU)/leucovorin (LV)with FU/LValone in patients with metastatic colorectal cancer. J Clin Oncol 2003;21:60–65.

9. Hurwitz H, Fehrenbacher L, Cartwright T, Hainsworth J, Hein W, Berlin J, Griffing S, Novotny W,Holmgren E, Kabbinavar F. Bevacizumab (a monoclonal antibody to vascular endothelial growth factor)prolongs survival in first-line colorectal cancer (CRC): Results of a phase III trial of bevacizumab incombination with bolus IFL (irinotecan, 5-FU-LV) as a first-line therapy in subjects with metastatic CRC.Proc Am Soc Clin Oncol; 22: Abstract 3645.

10. Hurwitz H, Fehrenbacher L, NovotnyW, Cartwright T, Hainsworth J, HeimW, Berlin J, Baron A, GriffingS, Holmgren E, Ferrara N, Fyfe G, Rogers B, Ross R, Kabbinavar F. Bevacizumab plus irinotecan,fluorouracil, and leucovorin for metastatic colorectal cancer. N Engl J Med 2004;350:2335–2342.

11. Mitchell EP,Alberts SR, SchwartzMA,GiantonioBJ, Catalano PJ,MeropolNJ, O’Dwyer PJ, BensonAB.High-dose bevacizumab in combination with FOLFOX4 improves survival in patients with previouslytreated advanced colorectal cancer: Results from the Eastern Cooperative Oncology Group (ECOG) studyE3200. Gastrointestinal Cancers Symposium 2005; Abstract No: 169a.

12. GiantonioBJ, Catalano PJ,MeropolNJ, O’Dwyer PJ,Mitchell EP, Alberts SR, SchwartzMA, BensonAB.High-dose bevacizumab improves survival when combined with FOLFOX4 in previously treatedadvanced colorectal cancer: Results from the Eastern CooperativeOncologyGroup (ECOG) study E3200.J Clin Oncol (ASCO Meeting Abstracts) 2005;23:2.

13. Rini BI. VEGF-targeted therapy in metastatic renal cell carcinoma. Oncologist 2005;10:191–197.14. Yang JC, Haworth L, Sherry RM, Hwu P, Schwartzentruber DJ, Topalian SL, Steinberg SM, Chen HX,

Rosenberg SA. A randomized trial of bevacizumab, an anti-vascular endothelial growth factor antibody,for metastatic renal cancer. N Engl J Med 2003;349:427–434.

15. Hainsworth J, Sosman J, Spigel D, Schwert RC, Carrell DL, Hubbard F, Greco FA. Phase II trial ofbevacizumab and erlotinib in patients with metastatic renal carcinoma (RCC). J Clin Oncol (MeetingAbstracts) 2004;22:4502.

16. SpigelDR,Hainsworth JD, Sosman JA, RaefskyEL,MeluchAA, EdwardsD,Horowitz P, ThomasK,YostK, Stagg MP, Greco FA. Bevacizumab and erlotinib in the treatment of patients with metastatic renalcarcinoma (RCC): Update of a phase II multicenter trial. J Clin Oncol (Meeting Abstracts) 2005;23:4540.

17. Rini BI, Halabi S, Taylor J, Small EJ, Schilsky RL; Cancer and Leukemia Group B. 90206: A randomizedphase III trial of interferon-alpha or interferon-alpha plus anti-vascular endothelial growth factor antibody(bevacizumab) in metastatic renal cell carcinoma. Clin Cancer Res 2004;10:2584–2586.

18. Miller KD, Chap LI, Holmes FA, CobleighMA,MarcomPK, Fehrenbacher L, DicklerM,Overmoyer BA,Reimann JD, Sing AP, Langmuir V, Rugo HS. Randomized phase III trial of capecitabine compared withbevacizumab plus capecitabine in patients with previously treated metastatic breast cancer. J Clin Oncol2005;23:792–799.

19. Miller KD, Wang M, Gralow J, Dickler M, Cobleigh M, Perez EA, Shenker TN, Davidson NE. E2100: Arandomized phase III trial of paclitaxel plus bevacizumab as first-line therapy for locally recurrent ormetastatic breast cancer. Proc Am Soc Clin Oncol 2005. Late-breaking session.

20. Johnson DH, Fehrenbacher L, Novotny WF, Herbst RS, Nemunaitis JJ, Jablons DM, Langer CJ, DeVoreRF 3rd, Gaudreault J, Damico LA, Holmgren E, Kabbinavar F. Randomized phase II trial comparingbevacizumab plus carboplatin and paclitaxel with carboplatin and paclitaxel alone in previously untreatedlocally advanced or metastatic non-small-cell lung cancer. J Clin Oncol 2004;22:2184–2191.

ANTI-ANGIOGENIC DRUGS * 509

21. Herbst RS, Johnson DH, Mininberg E, Carbone DP, Henderson T, Kim ES, Blumenschein G, Jr., Lee JJ,Liu DD, Truong MT, HongWK, Tran H, Tsao A, Xie D, Ramies DA, Mass R, Seshagiri S, Eberhard DA,Kelley SK, Sandler A. Phase I/II trial evaluating the anti-vascular endothelial growth factor monoclonalantibody bevacizumab in combination with the HER-1/epidermal growth factor receptor tyrosine kinaseinhibitor erlotinib for patients with recurrent non-small-cell lung cancer. J Clin Oncol 2005;23:2544–2555.

22. Sandler AB, Gray R, Brahmer J, Dowlati A, Schiller JH, PerryMC, Johnson DH. Randomized phase II/IIItrial of paclitaxel (P) plus carboplatin (C) with or without bevacizumab (NSC #704865) in patients withadvanced non-squamous non-small cell lung cancer (NSCLC): An Eastern Cooperative Oncology Group(ECOG) Trial-E4599. J Clin Oncol (Meeting Abstracts) 2005;23:LBA4.

23. Barleon B, Reusch P, Totzke F, Herzog C, Keck C, Martiny-Baron G, Marme D. Soluble VEGFR-1secreted by endothelial cells and monocytes is present in human serum and plasma from healthy donors.Angiogenesis 2001;4:143–154.

24. Lin P, Polverini P, Dewhirst M, Shan S, Rao PS, Peters K. Inhibition of tumor angiogenesis using a solublereceptor establishes a role for Tie2 in pathologic vascular growth. J Clin Invest 1997;100:2072–2078.

25. Lin P, Sankar S, Shan S, Dewhirst MW, Polverini PJ, Quinn TQ, Peters KG. Inhibition of tumor growth bytargeting tumor endothelium using a soluble vascular endothelial growth factor receptor. Cell GrowthDiffer 1998;9:49–58.

26. Hoshida T, Sunamura M, Duda DG, Egawa S, Miyazaki S, Shineha R, Hamada H, Ohtani H, Satomi S,Matsuno S. Gene therapy for pancreatic cancer using an adenovirus vector encoding soluble flt-1 vascularendothelial growth factor receptor. Pancreas 2002;25:111–121.

27. Takayama K, Ueno H, Nakanishi Y, Sakamoto T, Inoue K, Shimizu K, Oohashi H, Hara N. Suppression oftumor angiogenesis and growth by gene transfer of a soluble form of vascular endothelial growth factorreceptor into a remote organ. Cancer Res 2000;60:2169–2177.

28. Ogawa T, Takayama K, Takakura N, Kitano S, Ueno H. Anti-tumor angiogenesis therapy using solublereceptors: Enhanced inhibition of tumor growth when soluble fibroblast growth factor receptor-1 is usedwith soluble vascular endothelial growth factor receptor. Cancer Gene Ther 2002;9:633–640.

29. Holash J, Davis S, Papadopoulos N, Croll SD, Ho L, Russell M, Boland P, Leidich R, Hylton D, Burova E,Ioffe E, Huang T, Radziejewski C, Bailey K, Fandl JP, Daly T, Wiegand SJ, Yancopoulos GD, Rudge JS.VEGF-Trap: AVEGF blocker with potent antitumor effects. Proc Natl Acad Sci USA 2002;99:11393–11398.

30. KimES, SerurA,Huang J,ManleyCA,McCruddenKW,Frischer JS, Soffer SZ,RingL,NewT,ZabiskyS,Rudge JS, Holash J, Yancopoulos GD,Kandel JJ, Yamashiro DJ. Potent VEGF blockade causes regressionof coopted vessels in a model of neuroblastoma. Proc Natl Acad Sci USA 2002;99:11399–11404.

31. Byrne AT, Ross L, Holash J, Nakanishi M, Hu L, Hofmann JI, Yancopoulos GD, Jaffe RB. Vascularendothelial growth factor-trap decreases tumor burden, inhibits ascites, and causes dramatic vascularremodeling in an ovarian cancer model. Clin Cancer Res 2003;9:5721–5728.

32. Huang J, Frischer JS, Serur A, KadenheA, Yokoi A,McCruddenKW,NewT, O’Toole K, Zabski S, RudgeJS, Holash J, YancopoulosGD,YamashiroDJ, Kandel JJ. Regression of established tumors andmetastasesby potent vascular endothelial growth factor blockade. Proc Natl Acad Sci USA 2003;100:7785–7790.

33. Saishin Y, Saishin Y, Takahashi K, Lima e Silva R, Hylton D, Rudge JS, Wiegand SJ, Campochiaro PA.VEGF-TRAP(R1R2) suppresses choroidal neovascularization and VEGF-induced breakdown of theblood-retinal barrier. J Cell Physiol 2003;195:241–248.

34. Dupont J, Rothenberg ML, Spriggs DR, Cedarbaum JM, Furfine ES, Cohen DP, Dancy I, Lee HS, CooperW, Lockhart AC. Safety and pharmacokinetics of intravenous VEGF Trap in a phase I clinical trial ofpatients with advanced solid tumors. J Clin Oncol (Meeting Abstracts) 2005;23:3029.

35. Liekens S, De Clercq E, Neyts J. Angiogenesis: regulators and clinical applications. Biochem Pharmacol2001;61:253–270.

36. Von Marschall Z, Scholz A, Cramer T, Schafer G, Schirner M, Oberg K, Wiedenmann B, Hocker M,Rosewicz S. Effects of interferon alpha onvascular endothelial growth factor gene transcription and tumorangiogenesis. J Natl Cancer Inst 2003;95:437–448.

37. Dubois J,HershonL, Carmant L,Belanger S, Leclerc JM,DavidM.Toxicity profile of interferon alfa-2b inchildren: A prospective evaluation. J Pediatr 1999;135:782–785.

38. Chang E, Boyd A, Nelson CC, Crowley D, Law T, Keough KM, Folkman J, Ezekowitz RA, Castle VP.Successful treatment of infantile hemangiomas with interferon-alpha-2b. J Pediatr Hematol Oncol1997;19:237–244.

39. Tamayo L, Ortiz DM, Orozco-Covarrubias L, Duran-McKinster C, Mora MA, Avila E, Teixeira F, Ruiz-Maldonado R. Therapeutic efficacy of interferon alfa-2b in infants with life-threatening gianthemangiomas. Arch Dermatol 1997;133:1567–1571.

510 * QUESADA, MUN‹ OZ-CHA¤ PULI, AND MEDINA

40. Kirkwood JM, Ibrahim JG, Sosman JA, Sondak VK, Agarwala SS, Ernstoff MS, Rao U. High-doseinterferon alfa-2b significantly prolongs relapse-free and overall survival compared with the GM2-KLH/QS-21 vaccine in patients with resected stage IIB-III melanoma: Results of intergroup trial E1694/S9512/C509801. J Clin Oncol 2001;19:2370–2380.

41. Kilbridge KL, Cole BF, Kirkwood JM, Haluska FG, Atkins MA, Ruckdeschel JC, Sock DE, Nease RF,Weeks JC. Quality-of-life—Adjusted survival analysis of high-dose adjuvant interferon alfa-2b for high-risk melanoma patients using intergroup clinical trial data. J Clin Oncol 2002;20:1311–1318.

42. Kirkwood JM, Bender C, Agarwala S, Tarhini A, Shipe-Spotloe J, Smelko B, Donnelly S, Stover L.Mechanisms and management of toxicities associated with high-dose interferon alfa-2b therapy. J ClinOncol 2002;20:3703–3718.

43. Kiss C, Kiss M, Szegedi I, Arvai K, Toth J, Olah E. Interferon-alpha therapy in children with malignantdiseases: Clinical experience in twenty-four patients treated in a single pediatric oncology unit. MedPediatr Oncol 2002;39:115–119.

44. LensMB, DawesM. Interferon alfa therapy for malignant melanoma: A systematic review of randomizedcontrolled trials. J Clin Oncol 2002;20:1818–1825.

45. Hauschild A, Weichenthal M, Balda BR, Becker JC, Wolff HH, Tilgen W, Schulte KW, Ring J,Schadendorf D, Lischner S, Burg G, Dummer R. Prospective randomized trial of interferon alfa-2b andinterleukin-2 as adjuvant treatment for resected intermediate- and high-risk primary melanoma withoutclinically detectable node metastasis. J Clin Oncol 2003;21:2883–2888.

46. Hancock BW, Wheatley K, Harris S, Ives N, Harrison G, Horsman JM, Middleton MR, Thatcher N,LoriganPC,Marsden JR,BurrowsL,GoreM.Adjuvant interferon in high-riskmelanoma: TheAIMHIGHStudy—United Kingdom Coordinating Committee on Cancer Research Randomized Study of AdjuvantLow-Dose Extended-Duration Interferon Alfa-2a in High-Risk Resected Malignant Melanoma. J ClinOncol 2004;22:53–61.

47. Kirkwood JM, Manola J, Ibrahim J, Sondak V, Ernstoff MS, Rao U. A pooled analysis of easterncooperative oncology group and intergroup trials of adjuvant high-dose interferon for melanoma. ClinCancer Res 2004;10:1670–1677.

48. Moschos SJ, Kirkwood JM, Konstantinopoulos PA. Present status and future prospects for adjuvanttherapy of melanoma: Time to build upon the foundation of high-dose interferon alfa-2b. J Clin Oncol2004;22:11–14.

49. Sunkara U,Walczak JR, Summerson L, Rogers T, EisenbergerM, Denmeade S, Pili R, Huff CA, SinibaldiV, Carducci MA. A phase II trial of temozolomide and IFN-alpha in patients with advanced renal cellcarcinoma. J Interferon Cytokine Res 2004;24:37–41.

50. Schuchter LM. Adjuvant interferon therapy for melanoma: High-dose, low-dose, no dose, which dose?. JClin Oncol. 2004;22:7–10.

51. Hosang M. Suramin binds to platelet-derived growth factor and inhibits its biological activity. J CellBiochem 1985;29:265–273.

52. Danesi R, Del BS, Soldani P, Campagni A, La RR, Myers CE, Paparelli A, Del TM. Suramin inhibitsbFGF-induced endothelial cell proliferation and angio-genesis in the chick chorioallantoicmembrane. Br JCancer 1993;68:932–938.

53. Middaugh CR, Mach H, Burke CJ, Volkin DB, Dabora JM, Tsai PK, Bruner MW, Ryan JA, Marfia KE.Nature of the interaction of growth factors with suramin. Biochemistry 1992;31:9016–9024.

54. Waltenberger J,MayrU, FrankH,HombachV. Suramin is a potent inhibitor of vascular endothelial growthfactor. A contribution to the molecular basis of its antiangiogenic action. J Mol Cell Cardiol1996;28:1523–1529.

55. Takano S, Gately S, Neville ME, Herblin WF, Gross JL, Engelhard H, Perricone M, Eidsvoog K, Brem S.Suramin, an anticancer and angiosuppressive agent, inhibits endothelial cell binding of basic fibroblastgrowth factor,migration, proliferation, and induction of urokinase-type plasminogen activator. CancerRes1994;54:2654–2660.

56. BocciG,Danesi R,Benelli U, Innocenti F,Di PaoloA, Fogli S,Del TaccaM. Inhibitory effect of suramin inrat models of angiogenesis in vitro and in vivo. Cancer Chemother Pharmacol 1999;43:205–212.

57. Pesenti E, Sola F, Mongelli N, Grandi M, Spreafico F. Suramin prevents neovascularisation andtumour growth through blocking of basic fibroblast growth factor activity. Br J Cancer 1992;66:367–372.

58. Kaur M, Reed E, Sartor O, Dahut W, Figg WD. Suramin’s development: What did we learn? Invest NewDrugs 2002;20:209–219.

59. Miglietta L, Canobbio L, Granetto C, Vannozzi MO, Esposito M, Boccardo F. Suramin/epidoxorubicinassociation in hormone-refractory prostate cancer: Preliminary results of a pilot phase II study. J CancerRes Clin Oncol 1997;123:407–410.

ANTI-ANGIOGENIC DRUGS * 511

60. Mirza MR, Jakobsen E, Pfeiffer P, Lindebjerg-Clasen B, Bergh J, Rose C. Suramin in non-small cell lungcancer and advanced breast cancer. Two parallel phase II studies. Acta Oncol 1997;36:171–174.

61. Kaur M, Reed E, Sartor O, Dahut W, Figg WD. Suramin’s development: What did we learn? Invest NewDrugs 2002;20:209–219.

62. Vogelzang NJ, Karrison T, Stadler WM, Garcia J, Cohn H, Kugler J, Troeger T, Giannone L, Arrieta R,Ratain MJ, Vokes EE. A Phase II trial of suramin monthly�3 for hormone-refractory prostate carcinoma.Cancer 2004;100:65–71.

63. Braddock PS, Hu DE, Fan TP, Stratford IJ, Harris AL, Bicknell R. A structure-activity analysis ofantagonism of the growth factor and angiogenic activity of basic fibroblast growth factor by suramin andrelated polyanions. Br J Cancer 1994;69:890–898.

64. CiomeiM, PastoriW,MarianiM, Sola F,GrandiM,Mongelli N, LaRoccaRV, SteinCA,Danesi R,CooperMR, Uhrich M, Myers CE. New sulfonated distamycin A derivatives with bFGF complexing activity.Biochem Pharmacol 1994;47:295–302.

65. MeyersMO,GagliardiAR, FlattmannGJ, Su JL,WangYZ,WolteringEA. Suramin analogs inhibit humanangiogenesis in vitro. J Surg Res 2000;91:130–134.

66. Finch PW, Yee LK, Chu MY, Chen TM, Lipsky MH, Maciag T, Friedman S, Epstein MH, Calabresi P.Inhibition of growth factor mitogenicity and growth of tumor cell xenografts by a sulfonated distamycin Aderivative. Pharmacology 1997;55:269–278.

67. Schneider GP, Salcedo R, Dong HF, Kleinman HK, Oppenheim JJ, Howard OM. Suradista NSC 651016inhibits the angiogenic activity of CXCL12-stromal cell-derived factor 1alpha. Clin Cancer Res2002;8:3955–3960.

68. Groen HJ, de Vries EG,WynendaeleW, van der GraafWT, Fokkema E, LechugaMJ, Poggesi I, Dirix LY,van Oosterom AT. PNU-145156E, a novel angiogenesis inhibitor, in patients with solid tumors: A phase Iand pharmacokinetic study. Clin Cancer Res 2001;7:3928–3933.

69. Nguyen NM, Lehr JE, Pienta KJ. Pentosan inhibits angiogenesis in vitro and suppresses prostate tumorgrowth in vivo. Anticancer Res 1993;13:2143–2147.

70. Lush RM, Figg WD, Pluda JM, Bitton R, Headlee D, Kohler D, Reed E, Sartor O, Cooper MR. A phase Istudy of pentosan polysulfate sodium in patients with advanced malignancies. Ann Oncol 1996;7:939–944.

71. Marshall JL, Wellstein A, Rae J, DeLap RJ, Phipps K, Hanfelt J, Yunmbam MK, Sun JX, Duchin KL,Hawkins MJ. Phase I trial of orally administered pentosan polysulfate in patients with advanced cancer.Clin Cancer Res 1997;3:2347–2354.

72. Schwartsmann G, Sprinz E, Kalakun L, Yamagushi N, Sander E, Grivicich I, Koya R, Mans DR. Phase IIstudy of pentosan polysulfate (PPS) in patients with AIDS-related Kaposi’s sarcoma. Tumori1996;82:360–363.

73. Teichman JMH, The role of pentosan polysulfate in treatment approaches for interstitial cystitis. Rev Urol2002;4:S21–S27.

74. Ghosh P, Smith M. Osteoarthritis, genetic and molecular mechanisms. Biogerontology 2002;3:85–88.75. Arora N,Masood R, Zheng T, Cai J, Smith DL, Gill PS. Vascular endothelial growth factor chimeric toxin

is highly active against endothelial cells. Cancer Res 1999;59:183–188.76. Veenendaal LM, JinH,Ran S,CheungL,NavoneN,Marks JW,Waltenberger J, Thorpe P,RosenblumMG.

In vitro and in vivo studies of a VEGF121/rGelonin chimeric fusion toxin targeting the neovasculature ofsolid tumors Proc Natl Acad Sci USA 2002;99:7866–7871.

77. Frankel AE. Increased Sophistication of Immunotoxins. Clin Cancer Res 2002;8:942–944.78. Zhu Z, Witte L. Inhibition of tumor growth and metastasis by targeting tumor associated angiogenesis

with antagonists to the receptors of vascular endoithelial growth factor. Invest New Drugs 1999;17:195–212.

79. Usman N, Blatt LM. Nuclease-resistant synthetic ribozymes: Developing a new class of therapeutics. JClin Invest 2000;106:1197–1202.

80. Parry TJ, Cushman C, Gallegos AM, Agrawal AB, Richardson M, Andrews LE, Maloney L, Mokler VR,Wincott FE, Pavco PA.Bioactivity of anti-angiogenic ribozymes targeting Flt-1 andKDRmRNA.NucleicAcids Res 1999;27:2569–2577.

81. Parry TJ, Pavco PA, Sandberg JA. Ribozyme pharmacokinetic screening for predicting pharmacodynamicdosing regimens. In: Rossi J, Couture L, editors, Intracellular ribozyme applications: Principles andprotocols. Wymondham, UK: Horizon Scientific Press. 1999.

82. Pavco PA, Bouhana KS, Gallegos AM, Agrawal A, Blanchard KS, Grimm SL, Jensen KL, Andrews LE,Wincott FE, Pitot PA, Tressler RJ, Cushman C, Reynolds MA, Parry TJ. Antitumor and antimetastaticactivity of ribozymes targeting the mRNAof vascular growth factor receptors. Clinical Cancer Res2000;6:2094–2103.

512 * QUESADA, MUN‹ OZ-CHA¤ PULI, AND MEDINA

83. Sandberg JA, Sproul CD, Blanchard KS, Bellon L, Sweedler D, Powell JA, Caputo FA, Kornbrust DJ,Parker VP, Parry TJ, Blatt LM. Acute toxicology and pharmacokinetic assessment of a ribozyme(ANGIOZYME) targeting vascular endothelial growth factor receptor mRNA in the cynomolgus monkey.Antisense Nucleic Acid Drug Dev 2000;10:153–162.

84. Sandberg JA, BouhanaKS,GallegosAM,AgrawalAB, GrimmSL,Wincott FE, ReynoldsMA, Pavco PA,Parry TJ. Pharmacokinetics of an antiangiogenic ribozyme (ANGIOZYME) in the mouse. AntisenseNucleic Acid Drug Dev 1999;9:271–277.

85. Sandberg JA, Parker VP, Blanchard KS, Sweedler D, Powell JA, Kachensky A, Bellon L, Usman N,Rossing T, Borden E, Blatt LM. Pharmacokinetics and tolerability of an antiangiogenic ribozyme(ANGIOZYME) in healthy volunteers. J Clin Pharmacol 2000;40:1462–1469.

86. Weng DE, Usaman N. Angiozyme: A novel angiogenesis inhibitor. Curr Oncol Rep 2001;3:141–146.

87. Venook A, Hurwitz H, Cunningham C, Burris H, Aitchison R, Radka S, Pavco P, Capra W, Wolin M,Usman N. Relationship of clinical outcome in metastatic colorectal carcinoma to levels of solubleVEGFR-1: Results of a Phase II trial of ANGIOZYME, a ribozyme targeting the pre-mRNAofVEGFR-1,in combination with chemotherapy. Proc Am Soc Clin Oncol 2003;22:256, Abstract 1025.

88. FairbrotherWJ, Christinger HW, Cochran AG, Fuh G, Keenan CJ, Quan C, Shriver SK, Tom JY,Wells JA,Cunningham BC. Novel peptides selected to bind vascular endothelial growth factor target the receptor-binding site. Biochemistry 1998;37:17754–17764.

89. Hagedorn M, Bikfalvi A. Target molecules for anti-angiogenic therapy: From basic research to clinicaltrials. Crit Rev Oncol Hematol 2000;34:89–110.

90. PrewettM, Huber J, Li Y, SantiagoA,O’ConnorW,KingK,Overholser J, Hooper A, Pytowski B,Witte L,Bohlen P, Hicklin DJ. Antivascular endothelial growth factor receptor (fetal liver kinase 1) monoclonalantibody inhibits tumor angiogenesis and growth of several mouse and human tumors. Cancer Res1999;59:5209–5218.

91. Kunkel P, Ulbricht U, Bohlen P, Brockmann MA, Fillbrandt R, Stavrou D, Westphal M, Lamszus K.Inhibition of glioma angiogenesis and growth in vivo by systemic treatment with a monoclonal antibodyagainst vascular endothelial growth factor receptor-2. Cancer Res 2001;61:6624–6628.

92. Hicklin DJ, Witte L, Zhu Z, Liao F, Wu Y, Li Y, Bohlen P. Monoclonal antibody strategies to blockangiogenesis. Drug Disc Today 2001;6:517–528.

93. Zhu Z, Rockwell P, Lu D, Kotanides H, Pytowski B, Hicklin DJ, Bohlen P, Witte L. Inhibition of vascularendothelial growth factor-induced receptor activation with anti-kinase insert domain-containing receptorsingle-chain antibodies from a phage display library. Cancer Res 1998;58:3209–3214.

94. Posey JA, Ng TC, Yang B, Khazaeli MB, Carpenter MD, Fox F, Needle M, Waksal H, LoBuglio AF. Aphase I study of anti-kinase insert domain-containing receptor antibody, IMC-1C11, in patients with livermetastases from colorectal carcinoma. Clin Cancer Res 2003;9:1323–1332.

95. Lu D, Shen J, Vil MD, Zhang H, Jimenez X, Bohlen P, Witte L, Zhu Z. Tailoring in vitro selection for apicomolar affinity human antibody directed against vascular endothelial growth factor receptor 2 forenhanced neutralizing activity. J Biol Chem 2003;278:43496–43507.

96. Zhu Z, Hattori K, ZhangH, JimenezX, LudwigDL, Dias S, Kussie P, KooH,KimHJ, LuD, LiuM, TejadaR, Friedrich M, Bohlen P, Witte L, Rafii S. Inhibition of human leukemia in an animal model with humanantibodies directed against vascular endothelial growth factor receptor 2. Correlation between antibodyaffinity and biological activity. Leukemia 2003;17:604–611.

97. Zhu Z, Bohlen P, Witte L. Clinical development of angiogenesis inhibitors to vascular endothelial growthfactor and its receptors as cancer therapeutics. Current Cancer Drug Targets 2002;2:135–156.

98. Jimenez X, Lu D, Brennan L, Persaud K, Liu M, Miao H, Witte L, Zhu Z. A recombinant, fully human,bispecific antibody neutralizes the biological activities mediated by both vascular endothelial growthfactor receptors 2 and 3. Mol Cancer Ther 2005;4:427–434.

99. Fong TA, Shawver LK, Sun L, Tang C, App H, Powell TJ, Kim YH, Schreck R,Wang X, RisauW, UllrichA, Hirth KP, McMahon G. SU5416 is a potent and selective inhibitor of the vascular endothelial growthfactor receptor (Flk-1/KDR) that inhibits tyrosine kinase catalysis, tumor vascularization, and growth ofmultiple tumor types. Cancer Res 1999;59:99–106.

100. Shaheen RM, Davis DW, Liu W, Zebrowski BK, Wilson MR, Bucana CD, McConkey DJ, McMahon G,Ellis LM. Antiangiogenic therapy targeting the tyrosine kinase receptor for vascular endothelial growthfactor receptor inhibits the growth of colon cancer liver metastasis and induces tumor and endothelial cellapoptosis. Cancer Res 1999;59:5412–5416.

101. Angelov L, Salhia B, Roncari L, McMahon G, Guha A. Inhibition of angiogenesis by blocking activationof the vascular endothelial growth factor receptor 2 leads to decreased growth of neurogenic sarcomas.Cancer Res 1999;59:5536–5541.

ANTI-ANGIOGENIC DRUGS * 513

102. Schuuring J, Bussink J, Bernsen HJ, Peeters W, van Der Kogel AJ. Irradiation combined with SU5416:microvascular changes and growth delay in a human xenograft glioblastoma tumor line. Int J Radiat OncolBiol Phys 2005;61:529–534.

103. Mendel DB, Schreck RE,West DC, Li G, Strawn LM, Tanciongco SS, Vasile S, Shawver LK, CherringtonJM. The angiogenesis inhibitor SU5416 has long-lasting effects on vascular endothelial growth factorreceptor phosphorylation and function. Clin Cancer Res 2000;6:4848–4858.

104. Mendel DB, Laird AD, Smolich BD, Blake RA, Liang C, HannahAL, Shaheen RM, Ellis LM,Weitman S,Shawver LK, Cherrington JM. Development of SU5416, a selective small molecule inhibitor ofVEGF receptor tyrosine kinase activity, as an anti-angiogenesis agent. Anticancer Drug Des 2000;15:29–41.

105. Via LE, Gore-Langton RE, Pluda JM. Clinical trials referral resource. Current clinical trials administeringthe antiangiogenesis agent SU5416. Oncology (Williston Park) 2000;14:1312, 1315–1316, 1321–1323.

106. Eng C, Kindler HL, Stadler WM, Karczmar G, Heimann R, Wade JL, Nattam S, Agamah E, Vokes EE.SU5416 in advanced colorectal cancer (CRC): AUniversity of Chicago Phase II Consortium Study. ProcAm Soc Clin Oncol 2001;20:116b.

107. FiedlerWM, Tinnefeld H,Mende T, Gehling U, Vohwinkel G, Kelsey S, Scigalla P, Hossfeld DK. A phaseII study with SU5416 in patients with C-Kit positive AML. Proc Am Soc Clin Oncol 2001;20:288a.

108. Gajewski TF, Flickinger S, Heimann R, Karczmar G, Rini B, Dragovich T, Stadler WM. Phase II study ofthe anti-angiogenic Flk-1 tyrosine kinase inhibitor SU5416 inmelanoma: Initial results. ProcAmSoc ClinOncol 2001;20:360a.

109. Kindler HL,VogelzangNJ, ChienK, StadlerW,KarczmarG,HeimannR,Vokes EE. SU5416 inmalignantmesothelioma:AUniversity ofChicago phase II consortium study. ProcAmSocClinOncol 2001;20:341a.

110. Salzberg M, Pless M, Rochlitz C, Herrmann R, Cropp G, Hannah A, Scigalla P. A phase I andpharmacokinetic study of single oral administration of SU5416 in patients with advanced solid tumors.Proc Am Soc Clin Oncol 2001;20:98a.

111. Richard S, Croisille L, Yvart J, Casadeval N, Eschwege P, Aghakhani N, David P, Gaudric A, Scigalla P,Hermine O. Paradoxical secondary polycythemia in von Hippel-Lindau patients treated with anti-vascularendothelial growth factor receptor therapy. Blood 2002;99:3851–3853.

112. Giles FJ, Stopeck AT, Silverman LR, Lancet JE, CooperMA,HannahAL, Cherrington JM,O’Farrell AM,Yuen HA, Louie SG, HongW, Cortes JE, Verstovsek S, Albitar M, O’Brien SM, Kantarjian HM, Karp JE.SU5416, a small molecule tyrosine kinase receptor inhibitor, has biologic activity in patients withrefractory acute myeloid leukemia or myelodysplastic syndromes. Blood 2003;102:795–801.

113. KuenenBC,LeviM,Meijer JCM,KakkarAK, vanHinsberghVWM,Costeense PJ, PinedoHM,HoekmanK. Analysis of coagulation cascade and endotelial cell activation during inhibition of vascular endotelialgrowth factor/vascular endothelial growth factor receptor pathway in cancer patients. Arterioscler ThrombVasc Biol 2002;22:1500–1505.

114. Heymach JV, Desai J, Manola J, Davis DW, McConkey DJ, Harmon D, Ryan DP, Goss G, Quigley T, Vanden Abbeele AD, Silverman SG, Connors S, Folkman J, Fletcher CD, Demetri GD. Phase II study of theantiangiogenic agent SU5416 in patients with advanced soft tissue sarcomas. Clin Cancer Res 2004;10:5732–5740.

115. Giles FJ, Stopeck AT, Silverman LR, Lancet JE, CooperMA,HannahAL, Cherrington JM,O’Farrell AM,Yuen HA, Louie SG, HongW, Cortes JE, Verstovsek S, Albitar M, O’Brien SM, Kantarjian HM, Karp JE.SU5416, a small molecule tyrosine kinase receptor inhibitor, has biologic activity in patients withrefractory acute myeloid leukemia or myelodysplastic syndromes. Blood 2003;102:795–801.

116. ZangariM,Anaissie E, StopeckA,MorimotoA, TanN, Lancet J, CooperM,HannahA,Garcia-ManeroG,Faderl S, Kantarjian H, Cherrington J, Albitar M, Giles FJ. Phase II study of SU5416, a small moleculevascular endothelial growth factor tyrosine kinase receptor inhibitor, in patients with refractory multiplemyeloma. Clin Cancer Res 2004;10:88–95.

117. Fiedler W, Mesters R, Tinnefeld H, Loges S, Staib P, Duhrsen U, Flasshove M, Ottmann OG, Jung W,Cavalli F, Kuse R, Thomalla J, Serve H, O’Farrell AM, Jacobs M, Brega NM, Scigalla P, Hossfeld DK,BerdelWE. A phase 2 clinical study of SU5416 in patients with refractory acute myeloid leukemia. Blood2003;102:2763–2767.

118. Lara PN, Quinn DI, Margolin K, Meyers FJ, Longmate J, Frankel P, Mack PC, Turrell C, Valk P, Rao J,Buckley P, Wun T, Gosselin R, Galvin I, Gumerlock PH, Lenz HJ, Doroshow JH, Gandara DR. SU5416Plus interferon {alpha} in advanced renal cell carcinoma: A Phase II California Cancer ConsortiumStudy with Biological and Imaging Correlates of Angiogenesis Inhibition. Clin Cancer Res 2003;9:4772–4781.

119. Heymach JV, Desai J, Manola J, Davis DW, McConkey DJ, Harmon D, Ryan DP, Goss D, Quigley T, VandenAbbeele AD, Silverman SG, Connors S, Folkman J, Fletcher CDM,Demetri GD. Phase II Study of the

514 * QUESADA, MUN‹ OZ-CHA¤ PULI, AND MEDINA

antiangiogenic agent SU5416 in patients with advanced soft tissue sarcomas. Clin Cancer Res2004;10:5732–5740.

120. Peterson AC, Swiger S, Stadler WM, Medved M, Karczmar G, Gajewski TF. Phase II study of the Flk-1tyrosine kinase inhibitor SU5416 in advanced melanoma. Clin Cancer Res 2004;10:4048–4054.

121. StadlerWM, Cao D, Vogelzang NJ, Ryan CW, Hoving K,Wright R, Karrison T, Vokes EE. A randomizedPhase II trial of the antiangiogenic agent SU5416 in hormone-refractory prostate cancer. Clin Cancer Res2004;10:3365–3370.

122. Laird AD, Vajkoczy P, Shawver LK, Thurnher A, Liang C, Mohammadi M, Schessinger J, Ullrich A,Hubbard SR, Blake RA, Fong TA, Strawn LM, Sun L, Tang C, Hawtin R, Tang F, Shenoy N, Hirth KP,McMahon G, Cherrington JM. SU6668 is a potent antiangiogenic and antitumor agent that inducesregression of established tumors. Cancer Res 2000;60:4152–4160.

123. Laird AD, Christensen JG, Li G, Carver J, Smith K, Xin X,Moss KG, Louie SG,Mendel DB, CherringtonJM. SU6668 inhibits Flk-1/KDR and PDGFRbeta invivo, resulting in rapid apoptosis of tumor vasculatureand tumor regression in mice. FASEB J 2002;16:681–690.

124. Shaheen RM, Tseng WW, Davis DW, Liu W, Reinmuth N, Vellagas R, Wieczorek AA, Ogura Y,McConkey DJ, Drazan KE, Bucana CD, McMahon G, Ellis LM. Tyrosine kinase inhibition of multipleangiogenic growth factor receptors improves survival in mice bearing colon cancer liver metastases byinhibition of endothelial cell survival mechanisms. Cancer Res 2001;61:1464–1468.

125. Garofalo A, Naumova E, Manenti L, Ghilardi C, Ghisleni G, Caniatti M, Colombo T, Cherrington JM,Scanziani E, Nicoletti MI, Giavazzi R. The combination of the tyrosine kinase receptor inhibitor SU6668with paclitaxel affects ascites formation and tumor spread in ovarian carcinoma xenografts growingorthotopically. Clin Cancer Res 2003;9:3476–3485.

126. Fabbro D, Manley PW. Su-6668. Curr Opin Investig Drugs 2001;2:1142–1148.127. Rosen LS, Rosen PJ, Kabbinavar F, Rosen P, CroppG,HannahA. Phase I experiencewith SU6668, a novel

multiple receptor tyrosine kinase inhibitor in patients with advanced malignancies. Proc Am Soc ClinOncol 2001;20:97a.

128. Britten CD, Rosen LS, Kabbinavar F, Rosen P, Mulay M, Hernandez L, Brown J, Bello C, Kelsey SM,Scigalla P. Phase I trial of SU6668, a smallmolecule receptor tyrosine kinase inhibitor, given twice daily inpatients with advanced cancers. Proc Am Soc Clin Oncol 2002;21:28b.

129. Demetri GD, Desai J, Fletcher JA, Morgan JA, Fletcher CDM, Kazanovicz A, Van Den Abbeele A, BaumC,MakiR,HeinrichMC. SU11248, amulti-targeted tyrosine kinase inhibitor, can overcome imatinib (IM)resistance caused by diverse genomicmechanisms in patients (pts) withmetastatic gastrointestinal stromaltumor (GIST). J Clin Oncol 2004;22:3001.

130. Mendel DB, Laird AD, Xin X, Louie SG, Christensen JG, Li G, Schreck RE, Abrams TJ, Ngai TJ, Lee LB,Murray LJ, Carver J, Chan E,MoSKG, Haznedar JO, Sukbuntherng J, Blake RA, SunC, Tang C,Miller T,Shirazian S,McMahon G, Cherrington JM. In vivo antitumor activity of SU11248, a novel tyrosine kinaseinhibitor targeting vascular endothelial growth factor and platelet-derived growth factor receptors:Determination of a pharmacokinetic/pharmacodynamic relationship. Clin Cancer Res 2003;9:327–337.

131. O’Farrell AM,Abrams TJ, YuenHA,Ngai TJ, Louie SG,YeeKWH,WongLM,HongWM,Lee LB, TownA, Smolich BD, Manning WC, Murray LJ, Heinrich MC, Cherrington JM. SU11248 is a novel FLT3tyrosine kinase inhibitor with potent activity in vitro and in vivo. Blood 2003;101:3597–3605.

132. Abrams TJ, Murray LJ, Pesenti E, Holway VW, Colombo T, Lee LB, Cherrington JM, Pryer NK.Preclinical evaluation of the tyrosine kinase inhibitor SU11248 as a single agent and in combination with‘‘standard of care’’ therapeutic agents for the treatment of breast cancer. Mol Cancer Ther 2003;2:1011–1021.

133. Murray LJ, Abrams TJ, Long KR, Ngai TJ, Olson LM, Hong W, Keast PK, Brassard JA, O’Farrell AM,Cherrington JM, Pryer NK. SU11248 inhibits tumor growth and CSF-1R-dependent osteolysis in anexperimental breast cancer bone metastasis model. Clin Exp Metastasis 2003;20:757–766.

134. Schueneman AJ, Himmelfarb E, Geng L, Tan J, Donnelly E, Mendel D, McMahon G, Hallahan DE.SU11248 maintenance therapy prevents tumor regrowth after fractionated irradiation of murine tumormodels. Cancer Res 2003;63:4009–4016.

135. Yee KW, Schittenhelm M, O’Farrell AM, Town AR, McGreevey L, Bainbridge T, Cherrington JM,Heinrich MC. Synergistic effect of SU11248 with cytarabine or daunorubicin on FLT3 ITD-positiveleukemic cells. Blood 2004;104:4202–4209.

136. Fiedler W, Serve H, Dohner H, Schwittay M, Ottmann OG, O’Farrell AM, Bello CL, Allred R, ManningWC, Cherrington JM, Louie SG, HongW, Brega NM,Massimini G, Scigalla P, BerdelWE, Hossfeld DK.Aphase 1 study of SU11248 in the treatment of patientswith refractory or resistant acutemyeloid leukemia(AML) or not amenable to conventional therapy for the disease. Blood 2005;105:986–993.

ANTI-ANGIOGENIC DRUGS * 515

137. MotzerRJ, RiniBI,MichaelsonMD,RedmanBG,HudesGR,WildingG, FiglinRA,Zhu J,KimST,BaumC. SU011248, a novel tyrosine kinase inhibitor, shows antitumor activity in second-line therapy forpatients with metastatic renal cell carcinoma: Results of a phase 2 trial. J Clin Oncol (Meeting Abstracts)2004;22:4500.

138. Motzer RJ, Rini BI,MichaelsonMD,RedmanBG,HudesGR,WildingG, Bukowski RM,GeorgeDJ, KimST, Baum CM. Phase 2 trials of SU11248 show antitumor activity in second-line therapy for patients withmetastatic renal cell carcinoma (RCC). J Clin Oncol (Meeting Abstracts) 2005;23:4508.

139. Beaumont J, CellaD, Li J,Motzer R,Rini B,MichaelsonD,KimS,BaumC. Patient reported outcomes in aphase 2 trial of SU11248 for metastatic renal cell carcinoma. J Clin Oncol (Meeting Abstracts)2005;23:8167.

140. Demetri GD, George S, Heinrich MC, Fletcher JA, Fletcher CM, Desai J, Cohen DP, Scigalla P,Cherrington JM, Van Den Abbeele AD. Clinical activity and tolerability of the multi-targeted tyrosinekinase inhibitor SU11248 in patients (pts)withmetastatic gastrointestinal stromal tumor (GIST) refractoryto imatinib mesylate. Proc Am Soc Clin Oncol 2003;22: Abstract 3273.

141. Maki RG, Fletcher JA, Heinrich MC, Morgan JA, George S, Desai J, Scheu K, Fletcher CDM, Baum C,Demetri GD. Results from a continuation trial of SU11248 in patients (pts) with imatinib (IM)-resistantgastrointestinal stromal tumor (GIST). J Clin Oncol (Meeting Abstracts) 2005;23:9011.

142. Demetri GD, van Oosterom AT, Blackstein M, Garrett C, Shah M, Heinrich M, McArthur G, Judson I,Baum CM, Casali PG. Phase 3, multicenter, randomized, double-blind, placebo-controlled trial ofSU11248 in patients (pts) following failure of imatinib for metastatic GIST. J Clin Oncol (MeetingAbstracts) 2005;23:4000.

143. Miller KD, BursteinHJ, Elias AD, RugoHS,CobleighMA, PegramMD,Eisenberg PD, CollierM,AdamsBJ, Baum CM. Phase II study of SU11248, a multitargeted receptor tyrosine kinase inhibitor (TKI), inpatients (pts) with previously treated metastatic breast cancer (MBC). J Clin Oncol (Meeting Abstracts)2005;23:563.

144. KulkeM, Lenz HJ,Meropol NJ, Posey J, RyanDP, Picus J, Bergsland E, Stuart K, BaumCM, Fuchs CS. Aphase 2 study to evaluate the efficacy and safety of SU11248 in patients (pts) with unresectableneuroendocrine tumors (NETs). J Clin Oncol (Meeting Abstracts) 2005;23:4008.

145. Wood JM, Bold G, Buchdunger E, Cozens R, Ferrari S, Frei J, Hofmann F, Mestan J, Mett H, O’Reilly T,Persohn E, Rosel J, Schnell C, Stover D, Theuer A, Towbin H, Wenger F, Woods-Cook K, Menrad A,Siemeister G, Schirner M, Thierauch KH, Schneider MR, Drevs J, Martiny-Baron G, Totzke F. PTK787/ZK 222584, a novel and potent inhibitor of vascular endothelial growth factor receptor tyrosine kinases,impairs vascular endothelial growth factor-induced responses and tumor growth after oral administration.Cancer Res 2000;60:2178–2189.

146. Solorzano CC, Baker CH, Bruns CJ, Killion JJ, Ellis LM, Wood J, Fidler IJ. Inhibition of growth andmetastasis of human pancreatic cancer growing in nude mice by PTK 787/ZK222584, an inhibitor of thevascular endothelial growth factor receptor tyrosine kinases. Cancer Biother Radiopharm 2001;16:359–370.

147. Baker CH, SolorzanoCC, Fidler IJ. Blockade of vascular endothelial growth factor receptor and epidermalgrowth factor receptor signaling for therapy of metastatic human pancreatic cancer. Cancer Res 2002;62:1996–2003.

148. Drevs J, Mross K, Fuxius S. A phase-1 dose-escalating and pharmacokinetic (PK) study of the VEGF-receptor-inhibitor PTK787/ZK222584 (PTK/ZK) in patients with liver metastasis of advanced cancer.Proc Am Soc Clin Oncol 2001;20:100a.

149. Morgan B, Thomas AL, Drevs J, Hennig J, Buchert M, Jivan A, Horsfield MA, Mross K, Ball HA, Lee L,MietlowskiW, Fuxuis S, Unger C, O’Byrne K, Henry A, Cherryman GR, Laurent D, DuganM,Marme D,Steward WP. Dynamic contrast-enhanced magnetic resonance imaging as a biomarker for thepharmacological response of PTK787/ZK 222584, an inhibitor of the vascular endothelial growth factorreceptor tyrosine kinases, in patients with advanced colorectal cancer and liver metastases: Results fromtwo phase I studies. J Clin Oncol 2003;21:3955–3964.

150. StewardWP, ThomasA,MorganB,WiedenmannB, Bartel C, Vanhoefer U, Trarbach T, JunkerU, LaurentD, Lebwohl D. Expanded phase I/II study of PTK787/ZK 222584 (PTK/ZK), a novel, oral angiogenesisinhibitor, in combination with FOLFOX-4 as first-line treatment for patients with metastatic colorectalcancer. J Clin Oncol (Meeting Abstracts) 2004;22:259.

151. Hecht JR, Trarbach T, Jaeger E, Hainsworth J,Wolff R, LloydK, BodokyG, BornerM, Laurent D, JacquesC. A randomized, double-blind, placebo-controlled, phase III study in patients (Pts) with metastaticadenocarcinoma of the colon or rectum receiving first-line chemotherapy with oxaliplatin/5-fluorouracil/leucovorin and PTK787/ZK 222584 or placebo (CONFIRM-1). J Clin Oncol (Meeting Abstracts)2005;23:LBA3.

516 * QUESADA, MUN‹ OZ-CHA¤ PULI, AND MEDINA

152. ReardonD, FriedmanH, YungWKA,BradaM, Conrad C, Provenzale J, Jackson E, SerajuddinH, ChenB,Laurent D. A phase I/II trial of PTK787/ZK 222584 (PTK/ZK), a novel, oral angiogenesis inhibitor, incombination with either temozolomide or lomustine for patients with recurrent glioblastoma multiforme(GBM). J Clin Oncol (Meeting Abstracts) 2004;22:110.

153. Wedge SR,OgilvieDJ, DukesM,Kendrew J, Curwen JO, Hennequin LF, ThomasAP, Stokes ES, Curry B,Richmond GH, Wadsworth PF. ZD4190: An orally active inhibitor of vascular endothelial growth factorsignaling with broad-spectrum antitumor efficacy. Cancer Res 2000;60:970–975.

154. Wedge SR, Ogilvie DJ, Dukes M, Kendrew J, Chester R, Jackson JA, Boffey SJ, Valentine PJ,Curwen JO, Musgrove HL, Graham GA, Hughes GD, Thomas AP, Stokes ES, Curry B, Richmond GH,Wadsworth PF, Bigley AL, Hennequin LF. ZD6474 inhibits vascular endothelial growth factorsignaling, angiogenesis, and tumor growth following oral administration. Cancer Res 2002;62:4645–4655.

155. Williams KJ, Telfer BA, Brave S, Kendrew J, Whittaker L, Stratford IJ, Wedge SR. ZD6474, a potentinhibitor of vascular endothelial growth factor signaling, combined with radiotherapy: Schedule-dependent enhancement of antitumor activity. Clin Cancer Res 2004;10:8587–8593.

156. Ciardiello F, Caputo R, Damiano V, Caputo R, Troiani T, Vitagliano D, Carlomagno F, Veneziani BM,Fontanini G, Bianco AR, Tortora G. Antitumor effects of ZD6474, a small molecule vascular endothelialgrowth factor receptor tyrosine kinase inhibitor, with additional activity against epidermal growth factorreceptor tyrosine kinase. Clin Cancer Res 2003;9:1546–1556.

157. Taguchi F, Koh Y, Koizumi F, Tamura T, Saijo N, Nishio K. Anticancer effects of ZD6474, a VEGFreceptor tyrosine kinase inhibitor, in gefitinib (‘‘Iressa’’)-sensitive and resistant xenograft models. CancerSci 2004;95:984–989.

158. Minami H, Ebi H, Tahara M, Sasaki Y, Yamamoto N, Yamada Y, Tamura T, Saijo N. A phase I study of anoralVEGF receptor tyrosine kinase inhibitor ZD6474, in Japanese patientswith solid tumors. ProcAmSocClin Oncol 2003;22: Abstract 778.

159. Hurwitz H, Holden SN, Eckhardt SG, Rosenthal M, de Boer R, Rischin D, Green M, Basser R. Clinicalevaluation of ZD6474, an orally active inhibitor of VEGF signaling, in patientswith solid tumors. ProcAmSoc Clin Oncol 2002;22: Abstract 325.

160. Heymach JV, Johnson BE, Rowbottom JA, Fidias P, Lu C, Prager D, Roubec J, Csada E, Dimery I, HerbstRS. A randomized, placebo-controlled phase II trial of ZD6474 plus docetaxel, in patients with NSCLC. JClin Oncol (Meeting Abstracts) 2005;23:3023.

161. Johnson BE,Ma P,West H, Kerr R, Prager D, Sandler A, Herbst RS, Stewart DJ, Dimery IW, Heymach JV.Preliminary phase II safety evaluation of ZD6474, in combination with carboplatin and paclitaxel, as 1st-line treatment in patients with NSCLC. J Clin Oncol (Meeting Abstracts) 2005;23:7102.

162. Wilhelm S, Chien DS. BAY 43-9006: Preclinical data. Curr Pharm Des 2002;8:2255–2257.163. Lyons JF,Wilhelm S, Hibner B, Bollag G. Discovery of a novel Raf kinase inhibitor. Endocr Relat Cancer

2001;8:219–225.164. WilhelmSM,Carter C, TangL,Wilkie D,McNabolaA, RongH, ChenC, ZhangX,Vincent P,McHughM,

Cao Y, Shujath J, Gawlak S, Eveleigh D, Rowley B, Liu L, Adnane L, Lynch M, Auclair D, Taylor I,Gedrich R, Voznesensky A, Riedl B, Post LE, Bollag G, Trail PA. BAY 43-9006 exhibits broad spectrumoral antitumor activity and targets the RAF/MEK/ERK pathway and receptor tyrosine kinases involved intumor progression and angiogenesis. Cancer Res 2004;64:7099–7109.

165. Strumberg D, Voliotis D, Moeller JG, Hilger RA, Richly H, Kredtke S, Beling C, Scheulen ME, Seeber S.Results of phase I pharmacokinetic and pharmacodynamic studies of the Raf kinase inhibitor BAY 43-9006 in patients with solid tumours. Int J Clin Pharmacol Ther 2002;40:580–581.

166. Richly H, Kupsch P, Passage K, Grubert M, Hilger RA, Voigtmann R, Schwartz B, Brendel E, ChristensenO, Haase CG, Strumberg D. Results of a phase I trial of BAY 43-9006 in combination with doxorubicin inpatients with primary hepatic cancer. Int J Clin Pharmacol Ther 2004;42:650–651.

167. Awada A, Hendlisz A, Gil T, Bartholomeus S, Mano M, de Valeriola D, Strumberg D, Brendel E,Haase CG, Schwartz B, Piccart M. Phase I safety and pharmacokinetics of BAY 43-9006 administered for21 days on/7 days off in patients with advanced, refractory solid tumours. Br J Cancer 2005;92:1855–1861.

168. Flaherty KT, Redlinger M, Schuchter LM, Lathia CD, Weber BL, O’Dwyer PJ. Phase I/II,pharmacokinetic and pharmacodynamic trial of BAY43-9006 alone in patients withmetastaticmelanoma.J Clin Oncol (Meeting Abstracts) 2005;23:3037.

169. Rosner GL, Stadler W, Ratain MJ. Randomized discontinuation design: Application to cytostatic anti-neoplastic agents. J Clin Onc 2002;20:4478–4484.

170. Ratain MJ, Flaherty KT, Stadler WM, O’Dwyer P, Kaye S, Xiong H, Patnaik A, Gore M, Lee RJ, Eisen T.Preliminary antitumor activity of BAY 43-9006 in metastatic renal cell carcinoma and other advanced

ANTI-ANGIOGENIC DRUGS * 517

refractory solid tumors in a phase II randomized discontinuation trial (RDT). J Clin Oncol (MeetingAbstracts) 2004;22:4501.

171. Ratain MJ, Eisen T, Stadler WM, Flaherty KT, Gore M, Desai A, Patnaik A, Xiong HQ, Schwartz B,O’Dwyer P. Final findings from a phase II, placebo-controlled, randomized discontinuation trial (RDT) ofsorafenib (BAY 43-9006) in patients with advanced renal cell carcinoma (RCC). J Clin Oncol (MeetingAbstracts) 2005;23:4544.

172. Escudier B, Szczylik C, Eisen T, StadlerWM, Schwartz B, ShanM, Bukowski RM. Randomized phase IIItrial of the Raf kinase and VEGFR inhibitor sorafenib (BAY 43-9006) in patients with advanced renal cellcarcinoma (RCC). J Clin Oncol (Meeting Abstracts) 2005;23:LBA4510.

173. Ciardiello F, TortoraG.A novel approach in the treatment of cancer: Targeting the epidermal growth factorreceptor. Clin Cancer Res 2001;7:2958–2970.

174. Kim ES, Khuri FR, Herbst RS. Epidermal growth factor receptor biology (IMC-C225). Curr Opin Oncol2001;13:506–513.

175. Luwor RB, Lu Y, Li X, Mendelsohn J, Fan Z. The antiepidermal growth factor receptor monoclonalantibody cetuximab/C225 reduces hypoxia-inducible factor-1 alpha, leading to transcriptional inhibitionof vascular endothelial growth factor expression. Oncogene 2005;24:4433–4441.

176. Cunningham D, Humblet Y, Siena S, Khayat D, Bleiberg H, Santoro A, Bets D, Mueser M, Harstrick A,Van CutsemE. 2003. Cetuximab (C225) alone or in combination with irinotecan (CPT-11) in patients withepidermal growth factor receptor (EGFR)-positive, irinotecan-refractory metastatic colorectal cancer.Proc Am Soc Clin Oncol 2002;22:252.

177. Chan AT, Hsu MM, Goh BC, Hui EP, Liu TW, Millward MJ, Hong RL, Whang-Peng J, Ma BB, To KF,Mueser M, Amellal N, Lin X, Chang AY. Multicenter, Phase II Study of cetuximab in combination withcarboplatin in patients with recurrent or metastatic nasopharyngeal carcinoma. J Clin Oncol 2005;23:3568–3576.

178. Gunnett K, Motzer R, Amato R, Todd M, Poo W-J, Cohen R, Baselga J, Cooper M, Robert F, Falcey J,Waksal H. Phase II study of anti-epidermal growth factor receptor (EGFr) antibody C225 alone in patients(pts) with metastatic renal cell carcinoma (RCC). Proc Am Soc Clin Oncol 1999;19:389.

179. Bonner JA, Ezekiel MP, Robert F, Meredith RF, Spencer SA, Waksal HW. Continued response followingtreatment with IMC-225, an EGFr MoAb, combined with RT in advanced head and neck malignancies.Proc Am Soc Clin Oncol 2000;19:4.

180. Hong WK, Arquette M, Nabell L, Needle MN, Waksal HW, Herbst RS. Efficacy and safety of the anti-epidermal growth factor receptor antibody (EGFR) IMC-C225 in combination with cisplatin in patientswith recurrent squamous cell carcinoma of the head and neck (SCCHN) refractory to cisplatin containingchemotherapy. Proc Am Soc Clin Oncol 2001;21:224.

181. Gatzemeier U, Pluzanska A, Szczesna A, Kaukel E, Roubec J, Brennscheidt U, De Rosa F,Mueller B, vonPawel J. Results of a phase III trial of erlotinib (OSI-774) combined with cisplatin and gemcitabine (GC)chemotherapy in advanced non-small cell lung cancer (NSCLC). J Clin Oncol (Meeting Abstracts)2004;22:617.

182. Van Laethem J-L, Raoul J-L, Mitry E, Brezault C, Husseini F, Cals L, Vedovato JC, Mueser MM, RougierP. Cetuximab (C225) in combination with bi-weekly irinotecan (CPT-11), infusional 5-fluorouracil (5-FU), and folinic acid (FA) in patients with metastatic colorectal cancer (CRC) expressing the epidermalgrowth factor receptor (EGFR). Preliminary safety and efficacy results. Proc Am Soc Clin Oncol 2003;22:264.

183. Abbruzzese JL, Rosenberg A, Xiong Q, LoBuglio A, Schmidt W, Wolff R, Needle M, Waksal H. Phase IIstudy of anti-epidermal growth factor receptor (EGFR) antibody cetuximab (IMC-C225) in combinationwith gemcitabine in patients with advanced pancreatic cancer. Proc Am Soc Clin Oncol 2001;21:130.

184. Bonner JA, Giralt J, Harari PM, Cohen R, Jones C, Sur RK, Rabin D, Azarnia N, Needle MN, Ang KK.Cetuximab prolongs survival in patients with locoregionally advanced squamous cell carcinoma of headand neck: A phase III study of high dose radiation therapy with or without cetuximab. J Clin Oncol(Meeting Abstracts) 2004;22:5507.

185. Polikoff J, Mitchell EP, Badarinath S, Graham CD, Jennis A, Chen TT, Gustafson TN, Langer C.Cetuximab plus FOLFOX for colorectal cancer (EXPLORE): Preliminary efficacy analysis of arandomized phase III trial. J Clin Oncol (Meeting Abstracts) 2005;23:3574.

186. Sobrero A, ScheithauerW,Maurel J, Mineur L, Fehrenbacher L, Kisker O, Kopit J, Langer C, SchlichtingM, Abubakr Y. Cetuximab plus irinotecan for metastatic colorectal cancer (mCRC): Safety analysis of thefirst 400 patients in a randomized phase III trial (EPIC). J Clin Oncol (Meeting Abstracts) 2005;23:3580.

187. LilenbaumR,Bonomi P,Ansari R,LynchT,GovindanR, JanneP,HannaN.Aphase II trial of cetuximab astherapy for recurrent non-small cell lung cancer (NSCLC): Final results. J Clin Oncol (Meeting Abstracts)2005;23:7036.

518 * QUESADA, MUN‹ OZ-CHA¤ PULI, AND MEDINA

188. Werner-Wasik M, Swann S, Curran W, Robert F, Komaki R, Lee CP, Jafar S, Share R, Choy H,Blumenschein G. A phase II study of cetuximab (C225) in combination with chemoradiation (CRT) inpatients (PTS)with stage IIIA/B non-small cell lung cancer (NSCLC):An interim overall toxicity report ofthe RTOG 0324 Trial. J Clin Oncol (Meeting Abstracts) 2005;23:7135.

189. Aghajanian C, Sabbatini P, Derosa F, Gerst S, Spriggs DR, Dupont J, Hensley ML, Pezzulli S, Konner J,Schilder RJ. A phase II study of cetuximab/paclitaxel/carboplatin for the initial treatment of advancedstage ovarian, primary peritoneal, and fallopian tube cancer. J Clin Oncol (Meeting Abstracts) 2005;23:5047.

190. TiseoM, LopreviteM,Ardizzoni A. Epidermal growth factor receptor inhibitors: A new prospective in thetreatment of lung cancer. Curr Med Chem Anti-Canc Agents 2004;4:139–148.

191. Cella D,Natale RB, LynchTJ, LeeC, CarboneD,Douillard JY,KayA,WolfM,HeyesA,Ward J. Disease-related symptoms in advanced non-small cell lung cancer (NSCLC) as measured by the Lung CancerSubscale (LCS) of the FACT-L questionnaire: Clinically meaningful improvements with gefitinib(IRESSA, ZD1839). Proc Am Soc Clin Oncol 2003;22:629.

192. Cho CD, Fisher GA, Halsey J, Jambalos CN, Advani RH, Wakelee H, Lum BL, Sikic BI. A II study ofgefitinib (ZD1839, IressaTM) combination with Folfox-4 (IFOX) in patients with unresectable ormetastatic colorectal cancer. Proc Am Soc Clin Oncol 2003;22:265.

193. Cortes-Funes H, Soto-Parra H. Extensive experience of disease control with gefitinib and the role ofprognostic markers. Br J Cancer 2003;89:S3–S8.

194. Cohen EE, Rosen F, Stadler WM, Recant W, Stenson K, Huo D, Vokes EE. Phase II trial of ZD1839 inrecurrent or metastatic squamous cell carcinoma of the head and neck. J Clin Oncol 2003;21:1980–1987.

195. Lieberman FS, Cloughesy T, DeAngelis L, Fine H, Fink K, Junck L, Kuhn J, Mehta M, Wen P, Prados M.Phase I-II study of ZD-1839 for recurrent malignant gliomas andmeningiomas progressing after radiationtherapy. Proc Am Soc Clin Oncol 2003;22: Abstract 421.

196. RansonM,Wardell S.Gefitinib, a novel, orally administered agent for the treatment of cancer. JClin PharmTher 2004;29:95–103.

197. Peery TS, Reardon DA, Quinn J, Ochs J, Wikstrand C, Stenzel TT, Bigner DD, Friedman HS, Rich JN,Dancey J. Phase II trial of ZD1839 (Iressa) for patients with first relapse glioblastoma. Proc Am Soc ClinOncol 2003;22: Abstract 396.

198. Robertson JFR, Gutteridge E, CheungK,Owers R, KoehlerM, Hamilton L, Gee J, Nicholson RI. Gefitinib(‘Iressa’, ZD1839) is active in acquired tamoxifen (TAM)-resistant oestrogen receptor (ER)-positive andER-negative breast cancer: Results from a Phase II study. Proc Am Soc Clin Oncol 2003;22: Abstract 23.

199. Janne PA, Gurubhagavatula S, Yeap BY, Lucca J, Ostler P, Skarin AT, Fidias P, Lynch TJ, Johnson BE.Outcomes of patientswith advanced non-small cell lung cancer treatedwith gefitinib (ZD1839, ‘Iressa’) onan expanded access study. Lung Cancer 2004;44:221–230.

200. Rich JN, ReardonDA, Peery T, Dowell JM,Quinn JA, PenneKL,WikstrandCJ, VanDuynLB,Dancey JE,McLendonRE,Kao JC, Stenzel TT,AhmedRasheed BK, Tourt-Uhlig SE, Herndon JE II, Vredenburgh JJ,Sampson JH, FriedmanAH, Bigner DD, FriedmanHS. Phase II trial of gefitinib in recurrent glioblastoma.J Clin Oncol 2004;22:133–142.

201. Miller VA, Kris MG, Shah N, Patel J, Azzoli C, Gomez J, Krug LM, Pao W, Rizvi N, Pizzo B, Tyson L,Venkatraman E, Ben-Porat L, Memoli N, Zakowski M, Rusch V, Heelan RT. Bronchioloalveolarpathologic subtype and smoking history predict sensitivity to gefitinib in advanced non-small-cell lungcancer. J Clin Oncol 2004;22:1103–1109.

202. LynchTJ, Bell DW,Sordella R,Gurubhagavatula S,OkimotoRA,BranniganBW,Harris PL,Haserlat SM,Supko JG, Haluska FG, Louis DN, Christiani DC, Settleman J, Haber DA. Activating mutations in theepidermal growth factor receptor underlying responsiveness of non-small-cell lung cancer to gefitinib. NEngl J Med 2004;350:2129–2139.

203. Paez JG, Janne PA, Lee JC, Tracy S, Greulich H, Gabriel S, Herman P, Kaye FJ, Lindeman N, Boggon TJ,Naoki K, Sasaki H, Fujii Y, Eck MJ, Sellers WR, Johnson BE, Meyerson M. EGFR mutations in lungcancer: Correlation with clinical response to gefitinib therapy. Science 2004;304:1497–500.

204. PaoW,MillerVA.Epidermal growth factor receptormutations, small-molecule kinase inhibitors, and non-small-cell lung cancer: Current knowledge and future directions. J Clin Oncol 2005;23:2556–2568.

205. Shepherd F, Pereira J, Ciuleanu TE, Tan EH, Hirsh V, Thongprasert S, Bezjak A, Tu D, Santabarbara P,Seymour L. A randomized placebo-controlled trial of erlotinib in patients with advanced non-small celllung cancer (NSCLC) following failure of 1st line or or 2nd line chemotherapy.ANational Cancer Instituteof Canada Clinical Trials Group (NCIC). J Clin Oncol (Meeting Abstracts) 2004;22:7022.

206. Moore MJ, Goldstein D, Hamm J, Kotecha J, Gallinger S, Au HJ, Nomikos D, Ding K, Ptaszynski M,Parulekar W. A Phase III double blind trial of gemcitabine � erlotinib in locally advanced/metastaticpancreatic carcinoma. ASCO Gastrointest Cancers Symp 2005; Abstract 77.

ANTI-ANGIOGENIC DRUGS * 519

207. MooreMJ, Goldstein D, Hamm J, Figer A, Hecht J, Gallinger S, Au H, Ding K, Christy-Bittel J, ParulekarW. Erlotinib plus gemcitabine compared to gemcitabine alone in patients with advanced pancreatic cancer.A phase III trial of the National Cancer Institute of Canada Clinical Trials Group [NCIC-CTG]. J ClinOncol (Meeting Abstracts) 2005;23:1.

208. Grunwald V, Hidalgo M. Development of the epidermal growth factor receptor inhibitor Tarceva (OSI-774). Adv Exp Med Biol 2003;532:235–246.

209. Sandler A. Clinical experience with the HER1/EGFR tyrosine kinase inhibitor erlotinib. Oncology2003;11:17–22.

210. Soulieres D, Senzer NN, Vokes EE, Hidalgo M, Agarwala SS, Siu LL. Multicenter phase II study oferlotinib, an oral epidermal growth factor receptor tyrosine kinase inhibitor, in patients with recurrent ormetastatic squamous cell cancer of the head and neck. J Clin Oncol 2004;22:77–85.

211. KimES,KiesM, Sabichi A, GinsbergL, Burke B,Holsinger C, Feng L, TruongMT,GlissonB, HongWK.Phase II study of combination cisplatin, docetaxel and erlotinib in patients with metastatic/recurrent headand neck squamous cell carcinoma (HNSCC). J Clin Oncol (Meeting Abstracts) 2005;23:5546.

212. Felip E, Rojo F, Heller A, Foernzler D, Valverde C, Klughammer B, Ramos J, Maacke H, Brennscheidt U,Baselga J. Phase II pharmacodynamic trial of erlotinib in advanced non-small cell lung cancer (NSCLC)patients previously treatedwith platinum-based chemotherapy: preliminary results. J ClinOncol (MeetingAbstracts) 2005;23:7100.

213. GiacconeG, Lechevalier T, ThatcherN, Smit E, JanmaatM,Rodriguez J,Oulid-AissaD, Soria JC.A phaseII study of erlotinib as first-line treatment of advanced non-small cell lung cancer. J Clin Oncol (MeetingAbstracts) 2005;23:7073.

214. Jackman D, Lucca J, Fidias P, Rabin MS, Lynch TJ, Ostler P, Skarin AT, Temel J, Johnson BE, Janne PA.Phase II study of the EGFR tyrosine kinase erlotinib in patients 70 years of age with previously untreatedadvanced non-small cell lung carcinoma. J Clin Oncol (Meeting Abstracts) 2005;23:7148.

215. Brewer CJ, Suh JH, Stevens GHJ, Barnett GH, Toms S, VogelbaumMA,Weil R, PeereboomDM. Phase IItrial of erlotinib with temozolomide and concurrent radiation therapy in patients with newly-diagnosedglioblastoma multiforme. J Clin Oncol (Meeting Abstracts) 2005;23:1567.

216. Cloughesy T, Yung A, Vrendenberg J, Aldape K, Eberhard D, Prados M, Vandenberg S, Klencke B,Mischel P. Phase II study of erlotinib in recurrent GBM: Molecular predictors of outcome. J Clin Oncol(Meeting Abstracts) 2005;23:1507.

217. Graham DL, Hillman DW, Hobday TJ, Rousey SR, Nair SG, Soori GS, Sabagh TM, Perez EA. N0234:Phase II study of erlotinib (OSI-774) plus gemcitabine as first-or second-line therapy for metastatic breastcancer (MBC). J Clin Oncol (Meeting Abstracts) 2005;23:644.

218. Spigel DR,Hainsworth JD, Sosman JA,RaefskyEL,MeluchAA, EdwardsD,Horowitz P, ThomasK,YostK, Stagg MP, Greco FA. Bevacizumab and erlotinib in the treatment of patients with metastatic renalcarcinoma (RCC): Update of a phase II multicenter trial. J Clin Oncol (Meeting Abstracts) 2005;23:4540.

219. Teicher BA,MenonK, Alvarez E, Galbreath E, Shih C, FaulMM.Antiangiogenic and antitumor effects ofa protein kinase C beta inhibitor in human HT-29 colon carcinoma and human CaKi1 renal cell carcinomaxenografts. Anticancer Res 2001;21:3175–3184.

220. Teicher BA,MenonK, Alvarez E, Galbreath E, Shih C, FaulMM.Antiangiogenic and antitumor effects ofa protein kinase C beta inhibitor in murine lewis lung carcinoma and human Calu-6 non-small-cell lungcarcinoma xenografts. Cancer Chemother Pharmacol 2001;48:473–480.

221. Teicher BA, Menon K, Alvarez E, Liu P, Shih C, Faul MM. Antiangiogenic and antitumor effects of aprotein kinase C beta inhibitor in human hepatocellular and gastric cancer xenografts. In Vivo2001;15:185–193.

222. TeicherBA,AlvarezE,MenonK,EstermanMA,ConsidineE, ShihC, FaulMM.Antiangiogenic effects ofa protein kinase C beta-selective small molecule. Cancer Chemother Pharmacol 2002;49:69–77.

223. Teicher BA, Menon K, Alvarez E, Shih C, Faul MM. Antiangiogenic and antitumor effects of a proteinkinase C beta inhibitor in human breast cancer and ovarian cancer xenografts. Invest New Drugs 2002;20:241–251.

224. Herbst RS, Thornton DE, Kies MS, Sinha V, Flanagan S, Cassidy CA, Carducci MA. Phase 1 study ofLY317615, a protein kinase C inhibitor (abstract 326). Proc AmAssoc Clin Oncol 2002;21: Abstract 326.

225. Ingber D, Fujita T, Kishimoto S, Sudo K, Kanamaru T, Brem H, Folkman J. Synthetic analogues offumagillin that inhibit angiogenesis and suppress tumour growth. Nature 1990;348:555–557.

226. Rodriguez-Nieto S, Medina MA, Quesada AR. A re-evaluation of fumagillin selectivity towardsendothelial cells. Anticancer Res 2001;21:3457–3460.

227. Abe J, Zhou W, Takuwa N, Taguchi J, Kurokawa K, Kumada M, Takuwa Y. A fumagillin derivativeangiogenesis inhibitor,AGM-1470, inhibits activation of cyclin-dependent kinases and phosphorylation of

520 * QUESADA, MUN‹ OZ-CHA¤ PULI, AND MEDINA

retinoblastoma gene product but not protein tyrosyl phosphorylation or protooncogene expression invascular endothelial cells. Cancer Res 1994;54:3407–3412.

228. Griffith EC, Su Z, Turk BE, Chen S, Chang YH, Wu Z, Biemann K, Liu JO. Methionine aminopeptidase(type 2) is the common target for angiogenesis inhibitors AGM-1470 and ovalicin. Chem Biol1997;4:461–471.

229. Bergers G, Javaherian K, Lo K.-M, Forkman J, Hanahan D. Effects of angiogenesis inhibitors onmultistage carcinogenesis in mice. Science 1999;284:808–812.

230. Lund EL, Bastholm L, Krisjansen PEG. Therapeutic synergy of TNP-470 and Ionizing radiation: Effectson tumor growth, vessel morphology, and angiogenesis in human glioblastoma multiforme xenographs.Clin Cancer Res 2000;6:971–978.

231. Satchi-Fainaro R, Puder M, Davies JW, Tran HT, Sampson DA, Greene AK, Corfas G, Folkman J.Targeting angiogenesis with a conjugate of HPMA copolymer and TNP-470. Nat Med 2004;10:255–261.

232. Kudelka AP, Levy T, Verschraegen CF, Edwards CL, Piamsomboon S, Termrungruanglert W, FreedmanRS,KaplanAL,KiebackDG,MeyersCA, JaeckleKA, Loyer E, StegerM,ManteR,Mavligit G,KillianA,TangRA,Gutterman JU,Kavanagh JJ. A phase I study of TNP-470 administered to patients with advancedsquamous cell cancer of the cervix. Clin Cancer Res 1997;3:1501–1505.

233. Dezube BJ, Von Roenn JH, Holden-Wiltse J, Cheung TW, Remick SC, Cooley TP, Moore J, SommadossiJP, Shriver SL, Suckow CW, Gill PS. Fumagillin analog in the treatment of Kaposi’s sarcoma: A phase IAIDS clinical trial group study. AIDS clinical trial groupNo. 215 team. J Clin Oncol 1998;16:1444–1449.

234. Bhargava P,Marshall JL, Rizvi N, DahutW, Yoe J, FigueraM, Phipps K, OngVS, Kato A, HawkinsMJ. Aphase I and pharmacokinetic study of TNP-470 administered weekly to patients with advanced cancer.Clin Cancer Res 1999;5:1989–1995.

235. Stadler WM, Kuzel T, Shapiro C, Sosman J, Clark J, Vogelzang NJ. Multi-institutional study of theangiogenesis inhibitor TNP-470 in metastatic renal carcinoma. J Clin Oncol 1999;17:2541–2545.

236. Herbst RS,Madden TL, TranHT, BlumenscheinGR,Meyers CA, Seabrooke LF,Khuri FR, Puduvalli VK,Allgood V, Fritsche HA, Hinton L, Newman RA, Crane EA, Fossella FV, Dordal M, Goodin T, HongWK.Safety and pharmacokinetic effects of TNP-470, an angiogenesis inhibitor, combined with paclitaxel inpatients with solid tumors: Evidence for activity in non-small-cell lung cancer. J Clin Oncol 2002;20:4440–4447.

237. SillsAK, Jr.,Williams JI, Tyler BM,EpsteinDS, SiposEP,Davis JD,McLaneMP, Pitchford S,CheshireK,Gannon FH, Kinney WA, Chao TL, Donowitz M, Laterra J, Zasloff M, Brem H. Squalamine inhibitsangiogenesis and solid tumor growth in vivo and perturbs embryonic vasculature. Cancer Res1998;58:2784–2792.

238. Akhter S, Nath SK, Tse CM, Williams J, Zasloff M, Donowitz M. Squalamine, a novel cationic steroid,specifically inhibits the brush-border Naþ/Hþ exchanger isoform NHE3. Am J Physiol 1999;276:C136–C144.

239. Bhargava P,Marshall JL, DahutW, Rizvi N, TrockyN,Williams JI, Hait H, Song S, Holroyd KJ, HawkinsMJ. A phase I and pharmacokinetic study of squalamine, a novel antiangiogenic agent, in patients withadvanced cancers. Clin Cancer Res 2001;7:3912–3919.

240. Herbst RS, Hammond LA, Carbone DP, Tran HT, Holroyd KJ, Desai A, Williams JI, Bekele BN, Hait H,Allgood V, Solomon S, Schiller JH. A phase I/IIA trial of continuous five-day infusion of squalaminelactate (MSI-1256F) plus carboplatin and paclitaxel in patients with advanced non-small cell lung cancer.Clin Cancer Res 2003;9:4108–4115.

241. GubaM, von Breitenbuch P, Steinbauer M, Koehl G, Flegel S, HornungM, Bruns CJ, Zuelke C, Farkas S,Anthuber M, Jauch KW, Geissler EK. Rapamycin inhibits primary and metastatic tumor growth byantiangiogenesis: involvement of vascular endothelial growth factor. Nat Med 2002;8:128–135.

242. Vignot S, Faivre S, Aguirre D, Raymond E.MTOR-targeted therapy of cancer with rapamycin derivatives.Annals Oncol 2005;16:525–537.

243. Schuler W, Sedrani R, Cottens S, Haberlin B, Schulz M, Schuurman HJ, Zenke G, Zerwes HG,Schreier MH. SDZ RAD, a new rapamycin derivative: pharmacological properties in vitro and in vivo.Transplantation 1997;64:36–42.

244. Huang S, Houghton PJ. Inhibitors of mammalian target of rapamycin as novel antitumor agents: Frombench to clinic. Curr Opin Investig Drugs 2002;3:295–304.

245. Nashan B. Early clinical experience with a novel rapamycin derivative. Ther Drug Monit 2002;24:53–58.

246. Eisen HJ, Tuzcu EM, Dorent R, Kobashigawa J, Mancini D, Valantine-von Kaeppler HA, Starling RC,SorensenK,HummelM, Lind JM,AbeywickramaKH,Bernhardt P; RADB253StudyGroup. Everolimusfor the prevention of allograft rejection and vasculopathy in cardiac-transplant recipients. N Engl J Med2003;349:847–858.

ANTI-ANGIOGENIC DRUGS * 521

247. Sund M, Hamano Y, Sugimoto H, Sudhakar A, Soubasakos M, Yerramalla U, Benjamin LE, Lawler J,Kieran M, Shah A, Kalluri R. Function of endogenous inhibitors of angiogenesis as endothelium-specifictumor suppressors. Proc Natl Acad Sci USA 2005;102:2934–2939.

248. O’Reilly MS, Holmgren L, Shing Y, Chen C, Rosenthal RA, Moses M, Lane WS, Cao Y, Sage EH,Folkman J. Angiostatin: A novel angiogenesis inhibitor that mediates the suppression of metastases by aLewis lung carcinoma. Cell 1994;79:315–328.

249. O’ReillyMS, Boehm T, Shing Y, Fukai N, Vasios G, LaneWS, Flynn E, Birkhead JR, Olsen BR, FolkmanJ. Endostatin: An endogenous inhibitor of angiogenesis and tumor growth. Cell 1997;88:277–285.

250. DongZ,KumarR,YangX, Fidler IJ.Macrophage-derivedmetalloelastase is responsible for the generationof angiostatin in Lewis lung carcinoma. Cell 1997;88:801–810.

251. Gately S, Twardowski P, Stack MS, Patrick M, Boggio L, Cundiff DL, Schnaper HW, Madison L, VolpertO, Bouck N, Enghild J, Kwaan HC, Soff GA. Human prostate carcinoma cells express enzymatic activitythat converts human plasminogen to the angiogenesis inhibitor, angiostatin. Cancer Res 1996;56:4887–4890.

252. Sim BK. Angiostatin and endostatin: Endothelial cell-specific endogenous inhibitors of angiogenesis andtumor growth. Angiogenesis 1998;2:37–48.

253. Ren B, Wang Y, Ndebele K, Zhi Q, Chen FH, Wang YZ, Parangi S. Multiple signaling is involved inendostatin-mediated apoptosis in ECV 304 endothelial cells. Front Biosci 2005;10:1089–1097.

254. Twombly R. First clinical trials of endostatin yield lukewarm results. J Natl Cancer Inst 2002;94:1520–1521.

255. Herbst RS,HessKR, TranHT, Tseng JE,Mullani NA,Charnsangavej C,MaddenT,DavisDW,McConkeyDJ, O’Reilly MS, Ellis LM, Pluda J, Hong WK, Abbruzzese JL. Phase I study of recombinant humanendostatin in patients with advanced solid tumors. J Clin Oncol 2002;20:3792–3803.

256. Dresen A, Geiger P, Pluda J, Fogler W, Schiller JH, Wilding G. Phase I pharmacokinetic andpharmacodynamic study of recombinant human endostatin in patients with advanced solid tumors. J ClinOncol 2003;21:223–231.

257. Eder JP, Jr., Supko JG, Clark JW, Puchalski TA, Garcia-Carbonero R, Ryan DP, Shulman LN, Proper J,Kirvan M, Rattner B, Connors S, Keogan MT, Janicek MJ, Fogler WE, Schnipper L, Kinchla N, Sidor C,Phillips E, Folkman J, Kufe DW. Phase I clinical trial of recombinant human endostatin administered as ashort intravenous infusion repeated daily. J Clin Oncol 2002;20:3772–3784.

258. SunY,Wang J, Liu Y, SongX, ZhangY, Li K, ZhuY, ZhouQ, You L, YaoC. Results of phase III trial of rh-endostatin (YH-16) in advanced non-small cell lung cancer (NSCLC) patients. J Clin Oncol (MeetingAbstracts) 2005;23:7138.

259. De Moraes ED, Fogler WE, Grant D, Wahl M, Leeper D, Zrada S, Malin A, Connors S, Fortier AH,Dabrow M, Sidor C, Capizzi RL. Recombinant human angiostatin (rhA): A Phase I clinical trialassessing safety, pharmacokinetics (PK) and pharmacodynamics (PD). Proc Am Soc Clin Oncol2001;20:3.

260. Beerepoot LV,WitteveenED,GroenewegenG, FoglerWE, SimBK, SidarC, ZonnenbergBA, Schramel F,Gebbink MF, Voest EE. Recombinant human angiostatin by twice-daily subcutaneous injection inadvanced cancer: A pharmokinetic and long-term safety study. Clin Cancer Res 2003;9:4025–4033.

261. Folkman J. Role of angiogenesis in tumor growth and metastasis. Semin Oncol 2002;29:15–18.262. Kisker O, Becker CM, Prox D, Fannon M, D’Amato R, Flynn E, Fogler WE, Sim BK, Allred EN, Pirie-

Shepherd SR, Folkman J. Continuous administration of endostatin by intraperitoneally implantedosmoticpump improves the efficacy and potency of therapy in amouse xenograft tumormodel. Cancer Res2001;61:7669–7674.

263. Kim KS, Kim HS, Park JS, Kwon YG, Park YS. Inhibition of B16BL6 tumor progression bycoadministration of recombinant angiostatin K1-3 and endostatin genes with cationic liposomes. CancerGene Ther 2004;11:441–449.

264. Dutour A,Monteil J, Paraf F, Charissoux JL, Kaletta C, Sauer B, NaujoksK, RigaudM. Endostatin cDNA/Cationic liposome complexes as a promising therapy to prevent lungmetastases in osteosarcoma: Study ina human-like rat orthotopic tumor. Mol Ther 2005;11:311–319.

265. Coussens LM, Fingleton B, Matrisian LM. Matrix metalloproteinase inhibitors and cancer: Trials andtribulations. Science 2002;295:2387–2392.

266. Dove A. MMP inhibitors: Glimmers of hope amidst clinical failures. Nature Med 2002;8:95.267. Overall CM, Lopez-Otin C. Strategies forMMP inhibition in cancer: Innovations for the post-trial era. Nat

Rev Cancer 2002;2:657–672.268. Macaulay VM, O’Byrne KJ, Saunders MP, Braybrooke JP, Long L, Gleeson F, Mason CS, Harris AL,

BrownP, TalbotDC. Phase I study of intrapleural batimastat (BB-94), amatrixmetalloproteinase inhibitor,in the treatment of malignant pleural effusions. Clin Cancer Res 1999;5:513–520.

522 * QUESADA, MUN‹ OZ-CHA¤ PULI, AND MEDINA

269. Steward WP, Thomas AL. Marimastat: The clinical development of a matrix metalloproteinase inhibitor.Expert Opin Investig Drugs 2000;9:2913–2922.

270. Bramhall SR, Rosemurgy A, Brown PD, Bowry C, Buckels JA. Marimastat Pancreatic Cancer StudyGroup. Marimastat as first-line therapy for patients with unresectable pancreatic cancer: a randomizedtrial. J Clin Oncol 2001;19:3447–3455.

271. Shepherd FA, Giaccone G, Seymour L, Debruyne C, Bezjak A, Hirsh V, Smylie M, Rubin S, Martins H,Lamont A, Krzakowski M, Sadura A, Zee B. Prospective, randomized, double-blind, placebo-controlledtrial of marimastat after response to first-line chemotherapy in patients with small-cell lung cancer: A trialof the National Cancer Institute of Canada-Clinical Trials Group and the European Organization forResearch and Treatment of Cancer. J Clin Oncol 2002;20:4434–4439.

272. Sparano JA, Bernardo P, Stephenson P, Gradishar WJ, Ingle JN, Zucker S, Davidson NE. Randomizedphase III trial of marimastat versus placebo in patients with metastatic breast cancer who have respondingor stable disease after first-line chemotherapy: Eastern Cooperative Oncology Group trial E2196. J ClinOncol 2004;22:4683–4690.

273. ShalinskyDR, Brekken J, ZouH,McDermott CD, Forsyth P, Edwards D,Margosiak S, Bender S, Truitt G,Wood A, Varki NM, Appelt K. Broad antitumor and antiangiogenic activities of AG3340, a potent andselectiveMMP inhibitor undergoing advanced oncology clinical trials. Ann NYAcad Sci 1999;878:236–270.

274. ChantrainCF, ShimadaH, Jodele S,GroshenS,YeW,ShalinskyDR,WerbZ,CoussensLM,DeClerckYA.Stromal matrix metalloproteinase-9 (MMP-9) regulates the vascular architecture in neuroblastoma bypromoting pericyte recruitment. Cancer Res 2004;64:1675–1686.

275. D’Olimpio J, Hande K, Collier M, Michelson G, Paradiso L, Clendeninn N. Phase I study of the matrixmetalloproteinase inhibitor AG340 in combination with paclitaxel and carboplatin for the treatment ofpatients with advanced solid tumors. Proc Am Soc Clin Oncol 1999;18: Abstract 615.

276. Scatena R. Prinomastat, a hydroxamate-based matrix metalloproteinase inhibitor. A novel pharma-cological approach for tissue remodelling-related diseases. Expert Opin Investig Drugs 2000;9:2159–2165.

277. Bonomi P.Matrixmetalloproteinases andmatrixmetalloproteinase inhibitors in lung cancer. SeminOncol2002;29:78–86.

278. Bissett D, O’Byrne KJ, von Pawel J, Gatzemeier U, Price A, Nicolson M, Mercier R, Mazabel E, PenningC,ZhangMH,CollierMA,Shepherd FA. Phase III study ofmatrixmetalloproteinase inhibitor prinomastatin non-small-cell lung cancer. J Clin Oncol 2005;23:842–849.

279. Smylie M, Mercier R, Aboulafia D, Tucker R, Bonomi P, Collier M, Keller MR, Stuart-Smith J, KnowlesM, Clendeninn NJ, Shepherd F. Phase III study of the matrix metalloproteinases (MMP) inhibitorprinomastat in patients having advanced non-small cell lung cancer. Proc Am Soc Clin Oncol 2001;20:307a.

280. Bissett D, O’Bryne K, von Pawel J, Mercler R, Price A, Nicolson M, Shepherd F, Mazabel E, Penning C,Zhang MH, Collier MA. Phase III study of the matrix metalloprotease inhibitor prinomastat incombination with gemcitibine and cisplatin in non-small cell lung cancer. Proc Am Soc Clin Oncol 2002;21:296a.

281. Behrendt CE, Ruiz RB. Venous thromboembolism among patients with advanced lung cancer randomizedto prinomastat or placebo, plus chemotherapy. Thromb Haemost 2003;90:734–737.

282. Ferrario A, Chantrain CF, von Tiehl K, Buckley S, Rucker N, Shalinsky DR, Shimada H, DeClerck YA,Gomer CJ. The matrix metalloproteinase inhibitor prinomastat enhances photodynamic therapyresponsiveness in a mouse tumor model. Cancer Res 2004;64:2328–2332.

283. Ozerdem U, Mach-Hofacre B, Keefe K, Pham T, Soules K, Appelt K, Freeman WR. The effect ofprinomastat (AG3340), a synthetic inhibitor of matrix metalloproteinases, on posttraumatic proliferativevitreoretinopathy. Ophthalmic Res 2001;33:20–23.

284. Ozerdem U, Mach-Hofacre B, Varki N, Folberg R, Mueller AJ, Ochabski R, Pham T, Appelt K, FreemanWR. The effect of prinomastat (AG3340), a synthetic inhibitor of matrix metalloproteinases, on uvealmelanoma rabbit model. Curr Eye Res 2002;24:86–91.

285. Garcia C, Bartsch DU, Rivero ME, Hagedorn M, McDermott CD, Bergeron-Lynn G, Cheng L, Appelt K,Freeman WR. Efficacy of Prinomastat) (AG3340), a matrix metalloprotease inhibitor, in treatment ofretinal neovascularization. Curr Eye Res 2002;4:33–38.

286. Garcia C, Bartsch DU, Rivero ME, Hagedorn M, McDermott CD, Bergeron-Lynn G, Cheng L, Appelt K,Freeman WR. Efficacy of Prinomastat (AG3340), a matrix metalloprotease inhibitor, in treatment ofretinal neovascularization. Curr Eye Res 2002;24:33–38.

287. Rowinsky EK, Humphrey R, Hammond LA, Aylesworth C, Smetzer L, Hidalgo M, Morrow M, Smith L,Garner A, Sorensen JM, Von Hoff DD, Eckhardt SG. Phase I and pharmacologic of the specific matrix

ANTI-ANGIOGENIC DRUGS * 523

metalloprotease inhibitor BAY 12-9566 on a protracted oral daily dosing schedule in patients with solidmalignancies. J Clin Oncol 2000;18:178–186.

288. Heath EI, O’Reilly S, Humphrey R, Sundaresan P, Donehower RC, Sartorius S, Kennedy MJ, ArmstrongDK, Carducci MA, Sorensen JM, Kumor K, Kennedy S, Grochow LB. Phase I trial of the matrixmetalloproteinase inhibitor BAY12-9566 in patients with advanced solid tumors. Cancer ChemotherPharmacol 2001;48:269–274.

289. Bloomston M, Zervos EE, Rosemurgy AS. Matrix metalloproteinases and their role in pancreatic cancer:A review of preclinical studies and clinical trials. Ann Surg Oncol 2002;9:668–674.

290. Heath EI, O’Reilly S, Humphrey R, Sundaresan P, Donehower RC, Sartorius S, Kennedy MJ, ArmstrongDK, Carducci MA, Sorensen JM, Kumor K, Kennedy S, Grochow LB. Phase I trial of the matrixmetalloproteinase inhibitor BAY12-9566 in patients with advanced solid tumors. Cancer ChemotherPharmacol 2001;48:269–274.

291. DeplanqueG,Harris AL. Anti-angiogenic agents: Clinical trial design and therapies in development. Eur JCancer 2000;36:1713–1724.

292. Brown PD. Ongoing trials with matrix metalloproteinase inhibitors. Expert Opin Investig Drugs 2002;9:2167–2177.

293. Wall L, Talbot DC, Bradbury P, Jodrell DI. A phase I and pharmacological study of the matrixmetalloproteinase inhibitor BB-3644 in patients with solid tumours. Br J Cancer 2004;90:800–804.

294. Naglich JG, Jure-Kunkel M, Gupta E, Fargnoli J, Henderson AJ, Lewin AC, Talbott R, Baxter A, Bird J,Savopoulos R, Wills R, Kramer RA, Trail PA. Inhibition of angiogenesis and metastasis in two murinemodels by the matrix metalloproteinase inhibitor, BMS-275291. Cancer Res 2001;61:8480–8585.

295. LombardM,Wallace T, KubicekMF, Petzold GL,MitchellMA, Hendges SK,Wilks JW. Synthetic matrixmetalloproteinase inhibitors and tissue inhibitor of metalloproteinase (TIMP)-2, but not TIMP-1, inhibitshedding of tumor necrosis factor receptors in a human colon adenocarcinoma (Colo 205) cell line. CancerRes 1998;58:4001–4007.

296. RizviNA,Humphrey JS,Ness EA, JohnsonMD,GuptaE,WilliamsK,DalyDJ, SonnichsenD,ConwayD,Marshall J, Hurwitz H. A phase I study of oral BMS-275291, a novel nonhydroxamate sheddase-sparingmatrix metalloproteinase inhibitor, in patients with advanced or metastatic cancer. Clin Cancer Res 2004;10:1963–1970.

297. Hurwitz H, Humphrey J,WilliamsK, Ness E, ConwayA, Lockhart AC, BielefeldM, Tarby C, SonnichsenD, Gupta E, Huang MQ, Mao Y, Raphael M, Daley D, Marshall J, Johnson M, Rizvi N. A phase I trial ofBMS-275291: A novel non-hydrozamate, sheddase-sparing matrix metalloproteinase inhibitor with nodose-limiting arthritis. Proc Am Soc Clin Oncol 2001;20:98a.

298. Lara PN, Stadler WM, Longmate J, Quinn DI, Wexler J, Van Loan M, Twardowski P, Gumerlock PH,Vogelzang NJ, Vokes EE, Lenz HJ, Doroshow JH, Gandara DR. A randomized phase II trial of the matrixmetalloproteinase inhibitor BMS-275291 in hormone-refractory prostate cancer patients with bonemetastases. Clin Cancer Res 2006;12:1556–1563.

299. Poulaki V. BMS-275291. Bristol-Myers Squibb. Curr Opin Investig Drugs 2002;3:500–504.300. Douillard JY, Peschel C, Shepherd F, Paz-Ares L, Arnold A, Davis M, Tonato M, Smylie M, Tu D, Voi M,

Humphrey J, Ottaway J, Young K, Vreckem AV, Seymour L. Randomized phase II feasibility study ofcombining the matrix metalloproteinase inhibitor BMS-275291 with paclitaxel plus carboplatin inadvanced non-small cell lung cancer. Lung Cancer 2004;46:361–368.

301. Leighl NB, Paz-Ares L, Douillard JY, Peschel C, Arnold A, Depierre A, Santoro A, Betticher DC,Gatzemeier U, Jassem J, Crawford J, Tu D, Bezjak A. Randomized Phase III Study of MatrixMetalloproteinase Inhibitor BMS-275291 in Combination With Paclitaxel and Carboplatin in AdvancedNon-Small-Cell Lung Cancer: National Cancer Institute of Canada-Clinical Trials Group Study BR.18.,Humphrey JS, Voi M, Galbraith S, Hann K, Seymour L, Shepherd FA. J Clin Oncol 2005;23:2831–2839.

302. Miller KD, Saphner TJ,Waterhouse DM, Chen TT, Rush-Taylor A, Sparano JA,Wolff AC, CobleighMA,Galbraith S, SledgeGW.A randomized phase II feasibility trial ofBMS-275291 in patientswith early stagebreast cancer. Clin Cancer Res 2004;10:1971–1975.

303. Wood J, Schnell C, CozensR.CGS27023a, a potent and orally activematrixmetalloprotease inhibitorwithanti-tumour activity. Proc Am Assoc Cancer Res 1998;39:83A.

304. Nakamura ES, KoizumiK,Yamaura T, Saiki I. Anti-tumor angiogenic effect of amatrixmetalloproteinaseinhibitor MMI270. Anticancer Res 2003;23:411–417.

305. Levitt NC, Eskens FA, O’Byrne KJ, Propper DJ, Denis LJ, Owen SJ, Choi L, Foekens JA,Wilner S,WoodJM, NakajimaM, Talbot DC, StewardWP, Harris AL, Verweij J. Phase I and pharmacological study of theoral matrix metalloproteinase inhibitor, MMI270 (CGS27023A), in patients with advanced solid cancer.Clin Cancer Res 2001;7:1912–1922.

524 * QUESADA, MUN‹ OZ-CHA¤ PULI, AND MEDINA

306. Eatock M, Cassidy J, Johnson J, Morrison R, Devlin M, Blackey R, Owen S, Choi L, Twelves C.A dose-finding and pharmacokinetic study of the matrix metalloproteinase inhibitor MMI270(previously termed CGS27023A) with 5-FU and folinic acid. Cancer Chemother Pharmacol 2005;55:39–46.

307. Bramhall SR, Schulz J, Nemunaitis J, Brown PD, Baillet M, Buckels JA. A double-blind placebo-controlled, randomised study comparing gemcitabine and marimastat with gemcitabine and placebo asfirst line therapy in patients with advanced pancreatic cancer. Br J Cancer 2002;87:161–167.

308. Bramhall SR, Hallissey MT, Whiting J, Scholefield J, Tierney G, Stuart RC, Hawkins RE, McCulloch P,Maughan T, Brown PD, Baillet M, Fielding JW. Marimastat as maintenance therapy for patients withadvanced gastric cancer: A randomised trial. Br J Cancer 2002;86:1864–1870.

309. Groves MD, Puduvalli VK, Hess KR, Jaeckle KA, Peterson P, Yung WK, Levin VA. Phase II trial oftemozolomide plus the matrix metalloproteinase inhibitor, marimastat, in recurrent and progressiveglioblastoma multiforme. J Clin Oncol 2002;20:1383–1388.

310. Ramnath N, Creaven PJ. Matrix metalloproteinase inhibitors. Curr Oncol Rep 2004;6:96–102.311. Lokeshwar BL. MMP inhibition in prostate cancer. Ann N YAcad Sci 1999;878:271–289.312. SelzerMG, Zhu B, Block NL, Zhu B, Lokeshwar BL. CMT-3, a chemically modified tetracycline, inhibits

bonymetastases and delays the development of paraplegia in a rat model of prostate cancer. AnnNYAcadSci 1999;878:678–682.

313. Seftor RE, Seftor EA, De Larco JE, Kleiner DE, Leferson J, Stetler-StevensonWG,McNamara TF, GolubLM, Hendrix MJ. Chemically modified tetracyclines inhibit human melanoma cell invasion andmetastasis. Clin Exp Metastasis 1998;16:217–225.

314. Lokeshwar BL, Escatel E, Zhu B. Cytotoxic activity and inhibition of tumor cell invasion by derivatives ofa chemically modified tetracycline CMT-3 (COL-3). Curr Med Chem 2001;8:271–279.

315. LokeshwarBL, SelzerMG,ZhuBQ,BlockNL,GolubLM. Inhibition of cell proliferation, invasion, tumorgrowth and metastasis by an oral non-antimicrobial tetracycline analog (COL-3) in a metastatic prostatecancer model. Int J Cancer 2002;98:297–309.

316. Rudek MA, Figg WD, Dyer V, Dahut W, Turner ML, Steinberg SM, Liewehr DJ, Kohler DR, Pluda JM,ReedE. Phase I clinical trial of oral COL-3, amatrixmetalloproteinase inhibitor, in patientswith refractorymetastatic cancer. J Clin Oncol 2001;19:584–592.

317. Cianfrocca M, Cooley TP, Lee JY, Rudek MA, Scadden DT, Ratner L, Pluda JM, Figg WD, Krown SE,DezubeBJ.Matrixmetalloproteinase inhibitor COL-3 in the treatment ofAIDS-relatedKaposi’s sarcoma:A phase I AIDS malignancy consortium study. J Clin Oncol 2002;20:153–159.

318. Syed S, Takimoto C, Hidalgo M, Rizzo J, Kuhn JG, Hammond LA, Schwartz G, Tolcher A, Patnaik A,Eckhardt SG, RowinskyEK.A phase I and pharmacokinetic study of Col-3 (Metastat), an oral tetracyclinederivativewith potentmatrixmetalloproteinase and antitumor properties. ClinCancer Res 2004;10:6512–6521.

319. Gingras D, Renaud A, Mousseau N, Beliveau R. Shark cartilage extracts as antiangiogenic agents: Smartdrinks or bitter pills?. Cancer Metastasis Rev 2000;19:83–86.

320. GingrasD, BoivinD,Deckers C,Gendron S, Barthomeuf C,BeliveauR.Neovastat-a novel antiangiogenicdrug for cancer therapy. Anticancer Drugs 2003;14:91–96.

321. Boivin D, Gendron S, Beaulieu E, Gingras D, Beliveau R. The antiangiogenic agent Neovastat (AE-941)induces endothelial cell apoptosis. Mol Cancer Ther 2002;1:795–802.

322. Dupont E, Falardeau P, Mousa, Dimitriadou V, Pepin MC, Wang T, Alaoui-Jamali MA. Antiangiogenicand antimetastatic properties of Neovastat (AE-941), an orally active extract derived from cartilage tissue.Clin Exp Metast 2002;19:145–153.

323. Batist G, Patenaude F, Champagne P, Croteau D, Levinton C, Hariton C, Escudier B, Dupont E. Neovastat(AE-941) in refractory renal cell carcinoma patients: Report of a phase II trial with two dose levels. AnnOncol 2002;13:1259–1263.

324. Latreille J, Batist G, Laberge F, Champagne P, Croteau D, Falardeau P, Levinton C, Hariton C, EvansWK,Dupont E. Phase I/II trial of the safety and efficacy of AE-941 (Neovastat) in the treatment of non-small-cell lung cancer. Clin Lung Cancer 2003;4:231–236.

325. Falardeau P, Champagne P, Poyet P, Hariton C, Dupont E. Neovastat, a naturally occurringmultifunctionalantiangiogenic drug, in phase III clinical trials. Semin Oncol 2001;28:620–625.

326. Bukowski RM. AE-941, a multifunctional antiangiogenic compound: Trials in renal cell carcinoma.Expert Opin Investig Drugs 2003;12:1403–1411.

327. Dredge K. AE-941 (AEterna). Curr Opin Investig Drugs 2004;5:668–677.328. Escudier B, Venner P, Buckowski R, Szczylik C, Oudard S, Champagne P, Hariton C, Dupont E. Phase III

trial of neovastat in metastatic renal cell carcinoma patients refractory to immunotherapy. Proc Am SocClin Oncol 2003;22:211.

ANTI-ANGIOGENIC DRUGS * 525

329. Lu C, Komaki R, Herbst RS, Evans WK, Lee JJ, Truong M, Moore CA, Choy H, Bleyer A, Fisch MJ. Aphase III study of AE-941 with induction chemotherapy (IC) and concomitant chemoradiotherapy (CRT)for stage III non-small cell Lung cancer (NSCLC) (NCI T99–0046, RTOG 02–70, MDA 99–303): Aninterim report of toxicity and response. J Clin Oncol (Meeting Abstracts) 2005;23:7144.

330. Sauder DN, Dekoven J, Champagne P, Croteau D, Dupont E. Neovastat (AE-941), an inhibitor ofangiogenesis: Randomized phase I/II clinical trial results in patients with plaque psoriasis. J Am AcadDermatol 2002;47:535–541.

331. Thaloor D, Singh AK, Sidhu GS, Prasad PV, Kleinman HK, Maheswari RK. Inhibition of angiogenicdifferentiation of human umbilical endothelial cells by curcumin. Cell Growth Differ 1998;9:305–312.

332. Philip S, KunduGC.Osteopontin induces nuclear factor kappa B-mediated promatrixmetalloproteinase-2activation through I kappa B alpha/IKK signaling pathways, and curcumin (diferulolylmethane) down-regulates these pathways. J Biol Chem 2003;278:14487–14497.

333. ElkinM,MiaoHQ,Nagler A, Aingorn E, Reich R, Hemo I, DouHL, PinesM,Vlodavsky I. Halofuginone:A potent inhibitor of critical steps in angiogenesis progression. FASEB J 2000;14:2477–2485.

334. Rodriguez-Nieto S, Gonzalez-Iriarte M, Carmona R, Munoz-Chapuli R, Medina MA, Quesada AR.Antiangiogenic activity of aeroplysinin-1, a brominated compound isolated from amarine sponge. FASEBJ 2001. 10.1096/fj.01-0427fje, 2001a.

335. Brooks PC, Montgomery AMP, Rosenfeld M, Reisfeld RA, Hu T, Klier G, Cheresh DA. Integrin avb3antagonists promote tumor regression by inducing apoptosis of angiogenic blood vessels. Cell 1994;79:1157–1164.

336. Brooks PC, Stromblad S, Klemke R, Visscher D, Sarkar FH, Cheresh DA. Antiintegrin avb3 blocks humanbreast cancer growth and angiogenesis in human skin. J Clin Invest 1995;96:1815–1822.

337. Sipkins DA, Cheresh DA, Kazemi MR, Nevin LM, Bednarski MD, Li KC. Detection of tumorangiogenesis in vivo by avb3-targeted magnetic resonance imaging. Nat Med 1998;4:623–626.

338. Posey JA, KhazaeliMB,DelGrossoA, SalehMN, Lin CY,HuseW, LoBuglio AF.A pilot trial of Vitaxin, ahumanized anti-vitronectin receptor (anti avb3) antibody in patients with metastatic cancer. CancerBiother Radiopharm 2001;16:125–132.

339. Hersey P, Sosman J, O’Day S, Richards J, Bedikian A, Gonzalez R, Sharfman W, Weber R, Logan T,KirkwoodKM.Aphase II, randomized, open-label study evaluating the antitumor activity ofMEDI-522, ahumanizedmonoclonal antibody directed against the human alpha v beta 3 (avb3) integrin, � dacarbazine(DTIC) in patients with metastatic melanoma (MM). J Clin Oncol (Meeting Abstracts) 2005;23:7507.

340. Mikecz K. Vitaxin applied molecular evolution. Curr Opin Investig Drugs 2000;1:199–203.341. Patel SR, Jenkins J, Papadopolous N, Burgess MA, Plager C, Gutterman J, Benjamin RS. Pilot study of

vitaxin—an angiogenesis inhibitor—in patients with advanced leiomyosarcomas. Cancer 2001;92:1347–1348.

342. Wilder R. Integrin alpha v beta 3 as a target for treatment of rheumatoid arthritis and related rheumaticdiseases. Ann Rheum Dis 2002;61(Suppl 2):ii 96–99.

343. Mitjans F,Meyer T, Fittschen C, Goodman SL, Jonczyk A,Marshall JF, Reyes G, Piulats J. In vivo therapyof malignant melanoma by means of antagonists of v integrins. Int J Cancer 2000;87:716–723.

344. Burke PA, DeNardo SJ, Miers LA, Lamborn KR, Matzku S, DeNardo GL. Cilengitide targeting ofalpha(v)beta(3) integrin receptor synergizes with radioimmunotherapy to increase efficacy and apoptosisin breast cancer xenografts. Cancer Res 2002;62:4263–4272.

345. Haubner R, Gratias R, Diefenbach B, Goodman SL, Jonczyc A, Kessler H. Structural and functionalaspects of RGD containg cyclic pentapeptides as highly potent and selective integrin avb3 integrinantagonists. J Am Chem Soc 1996;118:7461–7472.

346. Dechantsreiter MA, Plankar E, Matha B, Lohof E, Holzemann G, Jonczyk A, Goodman SL, Kessler H. N-methylated cyclic RGD peptides as highly active and selective integrin avb3 integrin antagonists. J MedChem 1999;42:3033–3040.

347. Assa-Munt N, Jia X, Laakkonen P, Ruoslahti E. Solution structures and integrin binding activities of anRGD peptide with two isomers. Biochemistry 2001;40:2373–2378.

348. Eskens FA, Dumez H, Hoekstra R, Perschl A, Brindley C, Bottcher S, Wynendaele W, Drevs J, Verweij J,van Oosterom AT. Phase I and pharmacokinetic study of continuous twice weekly intravenousadministration of Cilengitide (EMD 121974), a novel inhibitor of the integrins alpha v beta 3 and alpha vbeta 5 in patients with advanced solid tumours. Eur J Cancer 2003;39:917–926.

349. Raguse JD, Gath HJ, Bier J, Riess H, Oettle H. Cilengitide (EMD 121974) arrests the growth of a heavilypretreated highly vascularised head and neck tumour. Oral Oncol 2004;40:228–230.

350. Kerr JS, Wexler RS, Mousa SA, Robinson CS, Wexler EJ, Mohamed S, Voss ME, Devenny JJ, CzerniakPM, Gudzelak A, Jr., Slee AM. Novel small molecule alpha v integrin antagonists: Comparative anti-cancer efficacy with known angiogenesis inhibitors. Anticancer Res 1999;19:959–968.

526 * QUESADA, MUN‹ OZ-CHA¤ PULI, AND MEDINA

351. Batt DG, Petraitis JJ, Houghton GC, Modi DP, Cain GA, Corjay MH, Mousa SA, Bouchard PJ, ForsytheMS, Harlow PP, Barbera FA, Spitz SM, Wexler RR, Jadhav PK. Disubstituted indazoles as potentantagonists of the integrin alpha(v)beta(3). J Med Chem 2000;43:41–58.

352. Miller WH, Keenan RM, Willette RN, Lark MW. Identificatuion and in vivo efficacy os small-moleculeantagonist of integrin avb3 (the vitronectin receptor). Drug Disc Today 2000;5:397–408.

353. Van Waes C, Enamorado-Ayala I, Hecht D, Sulica L, Chen Z, Batt DG, Mousa S. Effects of the novelalphav integrin antagonist SM256 and cis-platinum on growth of murine squamous cell carcinoma PAMLY8. Int J Oncol 2000;16:1189–1195.

354. Bourguet E, Baneres JL, Parello J, Lusinchi X, Girard JP, Vidal JP. Nonpeptide RGD antagonists: A novelclass of mimetics, the 5,8-disubstituted 1-azabicyclo[5.2.0] nonan-2-one lactam. Bioorg Med Chem Lett2003;13:1561–1564.

355. Hynes RO. A reevaluation of integrins as regulators of angiogenesis. Nat Med 2002;8:918–921.356. Tseng S, Pak G, Washenik K, Pomeranz MK, Shupack JL. Rediscovering thalidomide: A review of its

mechanism of action, side effects, and potential uses. J Am Acad Dermatol 1996;35:969–979.357. Sheskin J. Further observation with thalidomide in lepra reactions. Lepr Rev 1965;36:183–187.358. D’Amato RJ, Loughnan MS, Flynn E, Folkman J. Thalidomide is an inhibitor of angiogenesis. Proc Natl

Acad Sci USA 1994;91:4082–4085.359. Geitz H, Handt S, Zwingenberger K. Thalidomide selectively modulates the density of cell surface

molecules involved in the adhesion cascade. Immunopharmacol 1996;31:213–221.360. YabuT, TomimotoH,TaguchiY,YamaokaS, IgarashiY,Okazaki T. Thalidomide-induced anti-angiogenic

action is mediated by ceramide through depletion of VEGF receptors, and antagonized by sphingosine-1-phosphate. Blood 2005;106:125–134.

361. Singhal S, Mehta J, Desikan R, Ayers D, Roberson P, Eddlemon P, Munshi N, Anaissie E, Wilson C,Dhodapkar M, Zeddis J, Barlogie B. Antitumor activity of thalidomide in refractory multiple myeloma. NEngl J Med 1999;341:1565–1571.

362. Durie BG, Stepan DE. Efficacy of low dose thalidomide in multiple myeloma. Elec J Oncol 2000;1:1–8.

363. Barlogie B, Desikan R, Eddlemon P, Spencer T, Zeldis J, Munshi N, Badros A, Zangari M, Anaissie E,Epstein J, Shaughnessy J, Ayers D, Spoon D, Tricot G. Extended survival in advanced and refractorymultiple myeloma after single-agent thalidomide: Identification of prognostic factors in a phase 2 study of169 patients. Blood 2000;98:492–494.

364. Fine HA, Figg WD, Jaeckle K, Wen PY, Kyritsis AP, Loeffler JS, Levin VA, Black PM, Kaplan R, PludaJM, YungWK. Phase II trial of the antiangiogenic agent thalidomide in patients with recurrent high-gradegliomas. J Clin Oncol 2000;18:708–715.

365. Politi P, Reboredo G, Losso M, Vujacich C, Schwartsmann G, Lewi D. Phase I trial of thalidomide inAIDS-related Kaposi sarcoma. Proc Am Soc Clin Oncol 1999;18: Abstract 161.

366. Little RF, Wyvill KM, Pluda JM, Welles L, Marshall V, Figg WD, Newcomb FM, Tosato G, Feigal E,Steinberg SM, Whitby D, Goedert JJ, Yarchoan R. Activity of thalidomide in AIDS-related Kaposi’ssarcoma. J Clin Oncol 2000;18:2593–2602.

367. Dreicer R, Klein EA, Elson P, PeereboomD, Byzova T, Plow EF. Phase II trial of GM-CSFþ thalidomidein patients with androgen-independent metastatic prostate cancer. Urol Oncol 2005;23:82–86.

368. FiggWD, Arlen P, Gulley J, Fernandez P, NooneM, Fedenko K, HamiltonM, Parker C, Kruger EA, PludaJ, Dahut WL. A randomized phase II trial of docetaxel (taxotere) plus thalidomide in androgen-independent prostate cancer. Semin Oncol 2001;4:62–66.

369. Figg WD, Dahut W, Duray P, Hamilton M, Tompkins A, Steinberg SM, Jones E, Premkumar A, LinehanWM, FloeterMK, Chen CC, Dixon S, Kohler DR, Kruger EA, Gubish E, Pluda JM, Reed E. A randomizedphase II trial of thalidomide, an angiogenesis inhibitor, in patients with androgen-independent prostatecancer. Clin Cancer Res 2001;7:1888–1893.

370. Mesa RA, Steensma DP, Pardanani A, Li CY, Elliott M, Kaufmann SH, Wiseman G, Gray LA, SchroederG, Reeder T, Zeldis JB, Tefferi A. A phase 2 trial of combination low-dose thalidomide and prednisone forthe treatment of myelofibrosis with myeloid metaplasia. Blood 2003;101:2534–2541.

371. Gupta A, Cohen BH, Ruggieri P, Packer RJ, Phillips PC. Phase I study of thalidomide for the treatment ofplexiform neurofibroma in neurofibromatosis 1. Neurology 2003;60:130–132.

372. Reiriz AB, Richter MF, Fernandes S, Cancela AI, Costa TD, Di Leone LP, Schwartsmann G. Phase IIstudy of thalidomide in patients with metastatic malignant melanoma. Melanoma Res 2004;14:527–531.

373. Eisen T, Boshoff C, Mak I, Sapunar F, Vaughan MM, Pyle L, Johnston SR, Ahern R, Smith IE, Gore ME.Continuous low dose thalidomide: A phase II study in advanced melanoma, renal cell, ovarian and breastcancer. Br J Cancer 2000;82:812–817.

ANTI-ANGIOGENIC DRUGS * 527

374. Escudier B, LassauN, CouanetD,Angevin E,Mesrati F, Leborgne S, GarofanoA, Leboulaire C,DupouyN,Laplanche A. Phase II trial of thalidomide in renal-cell carcinoma. Ann Oncol 2002;13:1029–1035.

375. Motzer RJ, Berg W, Ginsberg M, Russo P, Vuky J, Yu R, Bacik J, Mazumdar M. Phase II trial ofthalidomide for patients with advanced renal cell carcinoma. J Clin Oncol 2002;20:302–306.

376. Baidas SM, Winer EP, Fleming GF, Harris L, Pluda JM, Crawford JG, Yamauchi H, Isaacs C, Hanfelt J,Tefft M, Flockhart D, Johnson MD, Hawkins MJ, Lippman ME, Hayes DF. Phase II evaluation ofthalidomide in patients with metastatic breast cancer. J Clin Oncol 2000;18:2710–2717.

377. Knisely JPS, Berkey BA, Chakravarti A, Yung WKA, Curran WJ, Robins HI, Movsas B, Brachman DG,Henderson RH, Mehta MP. RTOG 0118: A phase III study of conventional radiation therapy alone vs.conventional radiation therapy plus thalidomide for multiple brain metastases. J Clin Oncol (MeetingAbstracts) 2005;23:1500.

378. Alessandro R, Masiero L, Liotta LA, Kohn EC. The role of calcium in the regulation of invasion andangiogenesis. In Vivo 1996;10:153–160.

379. Alessandro R, Masiero LKL, Spoonster J, Kohn EC. Endothelial cell spreading on type IV collagen andspreading induced FAK phosphorylation is regulated by Caþþ influx. Biochem Biophys Res Commun1998;248:635–640.

380. Bauer KS, Cude KJ, Dixon SC, Kruger EA, Figg WD. Carboxyamido-triazole inhibits angiogenesis byblocking the calcium-mediated nitric-oxide synthase-vascular endothelial growth factor pathway. JPharmacol Exp Ther 2000;292:31–37.

381. Ge S, Rempel SA, Divine G, Mikkelsen T. Carboxyamido-triazole induces apoptosis in bovine aorticendothelial and human glioma cells. Clin Cancer Res 2000;6:1248–1254.

382. Kohn EC, JacobsW, KimYS, Alessandro R, Stetler-StevensonWG, Liotta LA. Calcium influxmodulatesexpression of matrix metalloproteinase-2 (72 kDa type IV collagenase, gelatinase A) expression. J BiolChem 1994;269:21505–21511.

383. Kohn EC, Alessandro R, Spoonster J, Wersto RP, Liotta LA. Angiogenesis: Role of calcium-mediatedsignal transduction. Proc Natl Acad Sci USA 1995;92:1307–1311.

384. Kohn EC, Reed E, Sarosy G, Christian M, Link CJ, Cole K, Figg WD, Davis PA, Jacob J, Goldspiel B,Liotta LA. Clinical investigation of a cytostatic calcium influx inhibitor in patients with refractory cancers.Cancer Res 1996;56:569–573.

385. Kohn EC, FiggWD, SarosyGA, BauerKS, Davis PA, SoltisMJ, ThompkinsA, Liotta LA, Reed E. Phase Itrial of micronized formulation carboxyamido-triazole in patients with refractory solid tumors:pharmacokinetics, clinical outcome and comparison of formulations. J Clin Oncol 1997;15:1985–1993.

386. Kohn EC, Reed E, Sarosy GA, Minasian L, Bauer KS, Bostick-Bruton F, Kulpa V, Fuse E, Tompkins A,Noone M, Goldspiel B, Pluda J, Figg WD, Liotta LA. A phase I trial of carboxyamido-triazole andpaclitaxel for relapsed solid tumors: Potential efficacy of the combination and demonstration ofpharmacokinetic interaction. Clin Cancer Res 2001;7:1600–1609.

387. Berlin J, Tutsch KD, Hutson P, Cleary J, Rago RP, Arzoomanian RZ, Alberti D, Feierabend C, Wildin G.Phase I clinical and pharmacokinetic study of oral carboxyamidotriazol, a signal transduction inhibitor. JClin Oncol 1997;15:781–789.

388. Bauer KS, FiggWD, Hamilton JM, Jones EC, Premkumar A, Steinberg SM, Dyer V, LinehanWM, PludaJM, Reed E. A pharmacokinetically guided phase II study of carboxyamido-triazole in androgen-independent prostate cancer. Clin Cancer Res 1999;5:2324–2329.

389. Nakashima T, Hirano S, Agata N, Kumagai H, Isshiki K, Yoshioka T, Ishizuka M, Maeda K, Takeuchi T.Inhibition of angiogenesis by a new isocoumarin, NM-3. J Antibiot (Tokyo) 1999;52:426–428.

390. Yin L, Ohno T,WeichselbaumR, Kharbanda S, KufeD. The novel isocoumarin 2-(8-hydroxy-6-methoxy-1-oxo-1H-2-benzopyran-3-yl) propionic acid (NM-3) induces lethality of human carcinoma cells bygeneration of reactive oxygen species. Mol Cancer Ther 2001;1:43–48.

391. Reimer CL, Agata N, Tammam JG, BambergM, DickersonWM, Kamphaus GD, Rook SL, Milhollen M,Fram R, Kalluri R, Kufe D, Kharbanda S. Antineoplastic effects of chemotherapeutic agents arepotentiated by NM-3, an inhibitor of angiogenesis. Cancer Res 2002;62:789–795.

392. Salloum RM, Jaskowiak NT, Mauceri HJ, Seetharam S, Beckett MA, Koons AM, Hari DM, Gupta VK,Reimer C, Kalluri R, Posner MC, Hellman S, Kufe DW, Weichselbaum RR. NM-3, an isocoumarin,increases the antitumor effects of radiotherapy without toxicity. Cancer Res 2000;60:6958–6963.

393. Kozin SV,BoucherY,HicklinDJ,Bohlen P, JainRK, SuitHD.Vascular endothelial growth factor receptor-2-blocking antibody potentiates radiation-induced long-term control of human tumor xenografts. CancerRes 2001;61:39–44.

394. Sakamoto T, Ishibashi T, Kimura H, Yoshikawa H, Spee C, Harris MS, Hinton DR, Ryan SJ. Effect oftecogalan sodium on angiogenesis in vitro by choroidal endothelial cells. Invest Ophthalmol Vis Sci1995;36:1076–1083.

528 * QUESADA, MUN‹ OZ-CHA¤ PULI, AND MEDINA

395. Tanaka NG, Sakamoto N, Inoue K, Korenaga H, Kadoya S, Ogawa H, Osada Y. Antitumor effects of anantiangiogenic polysaccharide from an Arthrobacter species with or without a steroid. Cancer Res1989;49:6727–6730.

396. YunmbamMK,WellsteinA.The bacterial polysaccharide tecogalan blocks growth of breast cancer cells invivo. Oncol Rep 2001;8:161–164.

397. Yoshikawa H, Ishibashi T, Hata Y, Inomata H, Sueishi K. The suppressive effect of tecogalan sodium onin vitro angiogenesis via the periendothelial proteolytic activities. Ophthalmic Res 2000;32:261–269.

398. Eckhardt SG, Burris HA, Eckardt JR, Weiss G, Rodriguez G, Rothenberg M, Rinaldi D, Barrington R,Kuhn JG,Masuo K, Sudo K, Atsumi R, Oguma T, Higashi L, Fields S, Smetzer L, Von Hoff DD. A phase Iclinical and pharmacokinetic study of the angiogenesis inhibitor, tecogalan sodium. Ann Oncol1996;7:491–496.

399. Miller KD, Sweeney CJ, Sledge GW Jr. Redefining the target: Chemotherapeutics as antiangiogenics. JClin Oncol 2001;19:1195–206.

400. Belani C, Lynch T. Docetaxel (Taxotere) in combination with platinums in patients with non-small celllung cancer: Trial data and implications for clinical management. Semin Oncol 2001;28:10–14.

401. Burris HA III. Docetaxel (Taxotere) plus trastuzumab (Herceptin) in breast cancer. Semin Oncol2001;28:38–44.

402. Calderoni A, Cerny T. Taxanes in lung cancer: A review with focus on the European experience. Crit RevOncol Hematol 2001;38:105–127.

403. Choy H. Taxanes in combined modality therapy for solid tumors. Crit Rev Oncol Hematol 2001;37:237–247.

404. Hortobagyi GN. Recent progress in the clinical development of docetaxel (Taxotere). Semin Oncol1999;26:32–36.

405. Rose WC, Fairchild C, Lee FY. Preclinical antitumor activity of two novel taxanes. Cancer ChemotherPharmacol 2001;47:97–105.

406. Milas L,MilasMM,MasonKA. Combination of taxanes with radiation: Preclinical studies. Semin RadiatOncol 1999;9:12–26.

407. Hennequin C, Favaudon V. Biological basis for chemo-radiotherapy interactions. Eur J Cancer 2002;38:223–230.

408. Herscher LL, Cook J. Taxanes as radiosensitizers for head and neck cancer. Curr Opin Oncol 1999;11:183–186.

409. Mason KA, Kishi K, Hunter N, Buchmiller L, Akimoto T, Komaki R, Milas L. Effect of docetaxel on thetherapeutic ratio of fractionated radiotherapy in vivo. Clin Cancer Res 1999;5:4191–4198.

410. Grant DS, Williams TL, Zahaczewsky M, Dicker AP. Comparison of antiangiogenic activities usingpaclitaxel (taxol) and docetaxel (taxotere). Int J Cancer 2003;104:121–129.

411. Kerbel RS. Tumor angiogenesis: Past, present and near future. Carcinogenesis 2000;21:505–515.412. Folkman J. Angiogenesis in cancer, vascular, rheumatoid and other disease. Nat Med 1995;1:27–31.413. Fox SB, Gasparini G, Harris AL. Angiogenesis: pathological, prognostic, and growth-factor pathways and

their link to trial design and anticancer drugs. Lancet Oncol 2001;2:278–289.414. McCarty MF, Liu W, Fan F, Parikh A, Reimuth N, Stoelzing O, Ellis LM. Promises and pitfalls of anti-

angiogenic therapy in clinical trials. Trends Mol Med 2003;9:5358.415. Cristofanilli M, Charnsangavej C, Hortobagyi GN. Angiogenesis modulation in cancer research: Novel

clinical approaches. Nat Rev Drug Discov 2002;1:415–426.416. DavisDW,McConjeyDJ,Abbruzzese JL,Herbst RS. Surrogatemarkers in antiangiogenesis clinical trials.

Br J Cancer 2003;89:8–14.417. Blagosklonny MV. Antiangiogenic therapy and tumor progression. Cancer Cell 2004;5:13–17.418. Inai T,MancusoM,HashizumeH,Baffert F, Haskell A, Baluk P,Hu-LoweDD, ShalinskyDR, ThurstonG,

Yancopoulos GD, McDonald DM. Inhibition of vascular endothelial growth factor (VEGF) signaling incancer causes loss of endothelial fenestrations, regression of tumor vessels, and appearance of basementmembrane ghosts. Am J Pathol 2004;165:35–52.

419. Vosseler S, Mirancea N, Bohlen P, Mueller MM, Fusenig NE. Angiogenesis inhibition by vascularendothelial growth factor receptor-2 blockade reduces stromal matrix metalloproteinase expression,normalizes stromal tissue, and reverts epithelial tumor phenotype in surface heterotransplants. Cancer Res2005;65:1294–1305.

420. Tong RT, Boucher Y, Kozin SV, Winkler F, Hicklin DJ, Jain RK. Vascular normalization by vascularendothelial growth factor receptor 2 blockade induces a pressure gradient across the vasculature andimproves drug penetration in tumors. Cancer Res 2004;64:3731–3736.

421. Jain RK. Normalization of tumor vasculature: An emerging concept in antiangiogenic therapy. Science2005;307:58–62.

ANTI-ANGIOGENIC DRUGS * 529

422. Winkler F, Kozin SV, Tong RT, Chae SS, Booth MF, Garkavtsev I, Xu L, Hicklin DJ, Fukumura D, diTomaso E, Munn LL, Jain RK. Kinetics of vascular normalization by VEGFR2 blockade governs braintumor response to radiation: Role of oxygenation, angiopoietin-1, and matrix metalloproteinases. CancerCell 2004;6:553–563.

423. Thorpe PE, Chaplin DJ, Blajey DC. The First International Conference on Vascular Targeting: Meetingoverview. Cancer Res 2003;63:1144–1147.

424. Tosetti F, Ferrari N, De Flora S, Albini A. Angioprevention: Angiogenesis is a common and key target forcancer chemopreventive agents. FASEB J 2002;16:2–14.

Ana R. Quesada is an Associate Professor of Biochemistry and Molecular Biology at University of Malaga,

Spain and Team Leader of a research group devoted to the search, screening, identification, and characterization

of new drugs with potential modulatory effects on angiogenesis.

RamonMunoz-Chapuli is a Full Professor of Animal Biology at University of Malaga, Spain and Team Leader

of a research group devoted to the study of the mechanisms involved in the control of cardiovascular

development, the embryonal origins of vascular and hematopoietic precursors, the epithelium-mesenchyme

transitions, and the evo–devo relationships of cardiovascular system.

Miguel Angel Medina is an Associate Professor of Biochemistry and Molecular Biology at University of

Malaga, Spain, and an active member of the research group leaded by ARQ. His research interests include

several aspects of basic oncology, including angiogenesis, but also amino acid and biogenic amine transport and

metabolism, as well as plasma membrane electron transport.

530 * QUESADA, MUN‹ OZ-CHA¤ PULI, AND MEDINA