molecular strategies targeting the host component of cancer to enhance tumor response to radiation...

9
doi:10.1016/j.ijrobp.2005.02.008 RTOG 2004 MOLECULAR STRATEGIES TARGETING THE HOST COMPONENT OF CANCER TO ENHANCE TUMOR RESPONSE TO RADIATION THERAPY DONG WOOK KIM, M.D., PH.D., JESSICA HUAMANI, M.S., ALLIE FU, B.S., AND DENNIS E. HALLAHAN, M.D. Department of Radiation Oncology, Vanderbilt Ingram Cancer Center, Nashville, TN The tumor microenvironment, in particular, the tumor vasculature, as an important target for the cytotoxic effects of radiation therapy is an established paradigm for cancer therapy. We review the evidence that the phosphoinositide 3-kinase (PI3K)/Akt pathway is activated in endothelial cells exposed to ionizing radiation (IR) and is a molecular target for the development of novel radiation sensitizing agents. On the basis of this premise, several promising preclinical studies that targeted the inhibition of the PI3K/Akt activation as a potential method of sensitizing the tumor vasculature to the cytotoxic effects of IR have been conducted. An innovative strategy to guide cytotoxic therapy in tumors treated with radiation and PI3K/Akt inhibitors is presented. The evidence supports a need for further investigation of combined-modality therapy that involves radiation therapy and inhibitors of PI3K/Akt pathway as a promising strategy for improving the treatment of patients with cancer. © 2006 Elsevier Inc. Akt, Vasculature targeted therapy, Endothelium, Radiation sensitizer, Combined modality therapy. INTRODUCTION Traditionally, ionizing radiation (IR) has been accepted as a therapeutic modality that directly targetes the deregulated cancer cells. Therefore, much attention to date has focused on agents that can lower radiation dose–response threshold (radiosensitizers), such as cisplatin or taxol (1). More re- cently, a new paradigm has been introduced that suggests that tumor host component (e.g., the tumor microvascula- ture) may also be a target for the cytotoxic effects of ionizing radiation (2–4). In particular, the tumor endothe- lium has received significant attention as a potential target for radiation sensitization. In fact, several preclinical studies have indicated the efficacy of combination of antiangio- genic agents and radiation therapy (RT) (5, 6). We and other investigators (7, 8) have focused on exploitation of novel mechanism that inhibit the phosphoinositide 3-kinase (PI3K)/Akt pathway as molecular targets for radiation sen- sitization of the host component of cancer. Endothelial cells, although sensitive to IR at higher doses (3, 4), are less sensitive to RT doses in the range of 2 to 3 Gy, in part because of activation of the PI3K/Akt cell viability signal- ing (9). Targeting this pathway in the tumor endothelium is an effective method of improving tumor response to frac- tionated RT in preclinical studies and is worthy of further clinical investigation (9 –13). We will briefly introduce an innovative potential strategy for guiding delivery of cyto- toxic agents to tumors that have received a combination of receptor tyrosine kinase (RTK) inhibitor and RT. PI3K/Akt PATHWAY Targeting radiation inducible cell viability pathway The PI3K/Akt pathway is known to be activated by growth-factor signaling through receptor tyrosine kinases, which can lead to activation of prosurvival pathways (14– 17). PI3K is composed of heterodimer of a p85 (relative molecular mass [Mr] (daltons) 85,000) adapter subunit and a p110 (Mr 100,000) catalytic subunit (18 –21). PI3K can be activated by a variety of RTKs (22–25). Furthermore, Akt, also referred to as protein kinase B, is activated when it is phosphorylated by phosphatidylinositide-dependent kinases 1 and 2 (PDK1 and PDK2) (26, 27), which are downstream of PI3K. Activated Akt has multiple downstream targets and is involved in regulation of apoptosis, glycolysis, and cell survival (Fig. 1)(28). PI3K/Akt activated by IR in endothelial cells PI3K/Akt-regulated signal transduction pathway through growth-factor signaling has been characterized as a viability pathway in many mammalian cells (28 –30). Ample evi- dence suggests that radiation could induce tumors to pro- duce angiogenic growth factors, which could be a mecha- Reprint requests to: Dennis E. Hallahan, M.D., Vanderbilt Univer- sity Medical Center, Department of Radiation Oncology, B-902, 1301 22nd Avenue South, Nashville, TN 37232-5671. Tel: (615) 343-9244; Fax: (615) 343-3075; E-mail: [email protected] Received Jan 4, 2005 and in revised form Feb 8, 2005. Accepted for publication Feb 8, 2005. Int. J. Radiation Oncology Biol. Phys., Vol. 64, No. 1, pp. 38 – 46, 2006 Copyright © 2006 Elsevier Inc. Printed in the USA. All rights reserved 0360-3016/06/$–see front matter 38

Upload: dong-wook-kim

Post on 21-Jun-2016

215 views

Category:

Documents


3 download

TRANSCRIPT

R

Ttco(cttilfhgim(sasbiatci

s2

Int. J. Radiation Oncology Biol. Phys., Vol. 64, No. 1, pp. 38–46, 2006Copyright © 2006 Elsevier Inc.

Printed in the USA. All rights reserved0360-3016/06/$–see front matter

doi:10.1016/j.ijrobp.2005.02.008

TOG 2004

MOLECULAR STRATEGIES TARGETING THE HOST COMPONENT OFCANCER TO ENHANCE TUMOR RESPONSE TO RADIATION THERAPY

DONG WOOK KIM, M.D., PH.D., JESSICA HUAMANI, M.S., ALLIE FU, B.S., AND

DENNIS E. HALLAHAN, M.D.

Department of Radiation Oncology, Vanderbilt Ingram Cancer Center, Nashville, TN

The tumor microenvironment, in particular, the tumor vasculature, as an important target for the cytotoxiceffects of radiation therapy is an established paradigm for cancer therapy. We review the evidence that thephosphoinositide 3-kinase (PI3K)/Akt pathway is activated in endothelial cells exposed to ionizing radiation (IR)and is a molecular target for the development of novel radiation sensitizing agents. On the basis of this premise,several promising preclinical studies that targeted the inhibition of the PI3K/Akt activation as a potential methodof sensitizing the tumor vasculature to the cytotoxic effects of IR have been conducted. An innovative strategy toguide cytotoxic therapy in tumors treated with radiation and PI3K/Akt inhibitors is presented. The evidencesupports a need for further investigation of combined-modality therapy that involves radiation therapy andinhibitors of PI3K/Akt pathway as a promising strategy for improving the treatment of patients with cancer.© 2006 Elsevier Inc.

Akt, Vasculature targeted therapy, Endothelium, Radiation sensitizer, Combined modality therapy.

tr

T

gw1maaap1ois

P

gpdd

F

INTRODUCTION

raditionally, ionizing radiation (IR) has been accepted as aherapeutic modality that directly targetes the deregulatedancer cells. Therefore, much attention to date has focusedn agents that can lower radiation dose–response thresholdradiosensitizers), such as cisplatin or taxol (1). More re-ently, a new paradigm has been introduced that suggestshat tumor host component (e.g., the tumor microvascula-ure) may also be a target for the cytotoxic effects ofonizing radiation (2–4). In particular, the tumor endothe-ium has received significant attention as a potential targetor radiation sensitization. In fact, several preclinical studiesave indicated the efficacy of combination of antiangio-enic agents and radiation therapy (RT) (5, 6). We and othernvestigators (7, 8) have focused on exploitation of novel

echanism that inhibit the phosphoinositide 3-kinasePI3K)/Akt pathway as molecular targets for radiation sen-itization of the host component of cancer. Endothelial cells,lthough sensitive to IR at higher doses (3, 4), are lessensitive to RT doses in the range of 2 to 3 Gy, in partecause of activation of the PI3K/Akt cell viability signal-ng (9). Targeting this pathway in the tumor endothelium isn effective method of improving tumor response to frac-ionated RT in preclinical studies and is worthy of furtherlinical investigation (9–13). We will briefly introduce annnovative potential strategy for guiding delivery of cyto-

Reprint requests to: Dennis E. Hallahan, M.D., Vanderbilt Univer-ity Medical Center, Department of Radiation Oncology, B-902, 1301

2nd Avenue South, Nashville, TN 37232-5671. Tel: (615) 343-9244; f

38

oxic agents to tumors that have received a combination ofeceptor tyrosine kinase (RTK) inhibitor and RT.

PI3K/Akt PATHWAY

argeting radiation inducible cell viability pathwayThe PI3K/Akt pathway is known to be activated by

rowth-factor signaling through receptor tyrosine kinases,hich can lead to activation of prosurvival pathways (14–7). PI3K is composed of heterodimer of a p85 (relativeolecular mass [Mr] (daltons) 85,000) adapter subunit andp110 (Mr 100,000) catalytic subunit (18–21). PI3K can bectivated by a variety of RTKs (22–25). Furthermore, Akt,lso referred to as protein kinase B, is activated when it ishosphorylated by phosphatidylinositide-dependent kinasesand 2 (PDK1 and PDK2) (26, 27), which are downstream

f PI3K. Activated Akt has multiple downstream targets ands involved in regulation of apoptosis, glycolysis, and cellurvival (Fig. 1) (28).

I3K/Akt activated by IR in endothelial cellsPI3K/Akt-regulated signal transduction pathway through

rowth-factor signaling has been characterized as a viabilityathway in many mammalian cells (28–30). Ample evi-ence suggests that radiation could induce tumors to pro-uce angiogenic growth factors, which could be a mecha-

ax: (615) 343-3075; E-mail: [email protected] Jan 4, 2005 and in revised form Feb 8, 2005. Accepted

or publication Feb 8, 2005.

nrhdgmhtytpmtrlvlPel4(Pedtblcl

iaa

Rr

tcgrbtltaFaieuacP

o

e artic

39Molecular strategies targeting the host component of cancer ● D. W. KIM et al.

ism of tumor’s paracrine regulation of endothelium’sesistance to radiation (6). However, several investigatorsave now demonstrated that human endothelial cells in vitroemonstrate activation of PI3K/Akt in the absence ofrowth-factor stimulus, which suggests an alternativeethod of this pathway’s activation (9, 31). Our laboratory

as demonstrated that increased doses of ionizing radiationreatment of endothelial cells leads to increased phosphor-lation of Akt, which plateaus at 3 Gy (9). Alternatively,reatment of HUVECs with 3 Gy led to increased Akthosphorylation within 5 minutes and maximally at 30inutes after treatment, without significant changes in the

otal Akt protein levels (9). Similar findings were alsoeported by Zingg et al. (31), who reported that in endothe-ial cells, PI3K/Akt activation occurs upon exposure to IRia upstream RTK activation, even in the absence of itsigand. The exact mechanism of how radiation activates theI3K/Akt pathway in the absence of growth factors inndothelial cell remains to be elucidated. Because endothe-ial cells, although susceptible to apoptosis at high doses (3,), are less sensitive to the lower doses of ionizing radiation2–3 Gy) (9), a reasonable hypothesis is that activation ofI3K/Akt prosurvival pathway by ionizing radiation (IR),ither through paracrine effect, or via mechanism indepen-ent of growth-factor stimulus, is in part responsible forumor vascular radioresistance. This hypothesis has noween validated in several preclinical studies from multipleaboratories that demonstrate inhibition of this pathwayonfers radiation sensitivity of tumor blood vessels even at

p

PDK1

PDK2

AKT P

P

GSK3β

FKHR

BAD

Cell

Survival

(Inhibits

Apoptosis)

Cell Cycle

Glucose Metabolism

P

P

P

Ionizing Radiation

Wortmannin

LY294002

Lithium

Fig. 1. Overview of signal-transduction pathway for PI3simplified view of radiation-mediated PI3K/Akt activatioare pharmaceutical compounds that target various levelsstudied in combination with radiation as described in th

ower doses of fractionated radiation (9–12, 31). The stud- p

es to date suggest at least 2 different methods of PI3K/Aktctivation in endothelial cells in response to ionizing radi-tion, which makes it an attractive target for therapy.

INHIBITION OF RADIATION-INDUCEDACTIVATION OF PI3K/Akt PATHWAY

eceptor tyrosine kinases as upstream targets foradiation sensitization of tumor vasculature

Receptor tyrosine kinases (RTKs) and their ligands par-icipate in angiogenesis, cell proliferation, and survival, andurrent data suggest that they are potential therapeutic tar-ets (32). Split-kinase domain RTKs such as platelet-de-ived growth factor (PDGF) receptor �, Flk-1/KDR, andasic fibroblast growth factor (bFGF) receptor play impor-ant roles in tumor angiogenesis. The antagonism of vascu-ar endothelial growth factor (VEGF) by antibodies (33) andhe use of Flk-1 receptor inhibitors has been shown to haventiangiogenic effects (11). Other RTK ligands, such asGF and PDGF, also appear to contribute to angiogenesisnd tumor growth (34). Moreover, bFGF has been shown tonhibit apoptosis in the microvasculature of mouse lungsxposed to radiation (35). FGF may also indirectly contrib-te to angiogenesis by up-regulation of VEGF (36). PDGFlso increases VEGF secretion in tumor cell lines (37). Thelinical relevance of these findings is that VEGF, FGF, andDGF are all up-regulated in response to radiation (8, 38).RTKs are key elements of the most well-studied families

f signaling pathways (39). VEGF receptor (VEGFR) is a

Receptor Tyrosine Kinase

wth Factors, RTK Ligands

mTOR

NF-κB

roliferation

ell Survival

Inhibits Apoptosis)

Cell Growth

Cell Survival

(Inhibits Apoptosis)

P

P

P

SU11248

SU5416

SU6668

PTK787

Rapamycin

RAD001

CCI-779

AP23573

IC486068

DC 101

activated by IR in tumor vasculature. This diagram is andothelial cells. In the yellow boxes, highlighted in red

PI3K/Akt signal transduction pathway that have beenle.

110

Gro

MDM2

P

C

(

p85

PI3K

K/Aktn in eof the

rototypical RTK for the vascular endothelial cells. The

prtipltpmtut

lbtpseVoVrrnegci

RtciatlrnSrooirSiSpaltgfT(i

Sweictccfcimmatsabp

tiPaKi5aPdcddtwditaS(sePdhfmttmdgabw

40 I. J. Radiation Oncology ● Biology ● Physics Volume 64, Number 1, 2006

hosphotyrosines in the RTK insert serve as binding sitesecognized by the SH2 domains of several signaling pro-eins, including PI3K, Grb2, and Shc (40, 41). This local-zed activity increases phosphatidylinositol-3,4,5-triphos-hate (PIP3) concentrations at the cell membrane, thusocally activating Akt. RTK signaling via its downstreamargets, which includes Akt, is thought to play a role inromoting cell proliferation and survival (Fig. 1). Further-ore, RTK signaling of tumor endothelium may contribute

o angiogenesis, maintenance of tumor vascular supply, and,ltimately, to tumor survival and resistance to cytotoxicherapy.

Because radiation activates PI3K/Akt signaling, severalaboratories have investigated the use of RTK inhibitors tolock the radiation induced activation of this pathway in theumor vasculature. Several RTK inhibitors are currently inreclinical and clinical studies, and a number of them havehown efficacy in tumor control in this setting (5). Forxample, treatment of tumors with combination of anti-EGF monoclonal antibody with radiation was based on thebservation that tumors treated with radiation inducedEGF production (8). In these studies, the paracrine up-

egulation of tumor angiogenesis that is induced by ionizingadiation was inhibited by blocking the activity of VEGF byeutralizing antibodies to this ligand. Multiple tumor mod-ls, such as Lewis lung, esophageal, squamous cell, andlioblastoma cells, exhibited greater than additive tumorontrol when anti-VEGF antibody was administered beforeonizing RT (8).

Several investigators have studied direct inhibition of theTK rather than its ligand as a method of improving cyto-

oxic therapy. Much of the effort has been directed atompounds that are either specific or broad-spectrum inhib-tors of VEGFR. Inhibition of these receptors can bechieved in 2 main ways: (1) an antibody directed againsthe extracellular domain of the receptor and (2) small mo-ecular compound specifically targeted for inhibition of theeceptor tyrosine kinases intracellularly (42). Examples ofarrow-spectrum inhibitors of VEGFR include DC101 andU5416. DC101 is a monoclonal antibody specifically di-ected against the murine VEGFR2. Kozin et al. (43) dem-nstrated that with DC101, they were able to lower the dosef radiation necessary to achieve 50% of tumor control, bothn radiation-sensitive (small-cell lung cancer) and in highlyesistant (glioblastoma multiforme) xenograft models.U5416 is a small-molecule receptor tyrosine kinase inhib-

tor. Geng et al. (11) demonstrated similarly the efficacy ofU5416 to revert the resistance to radiation-induced apo-tosis in HUVECs in vitro. Furthermore, combination ther-py with SU5416 and conventional fractionated doses of IRed to destruction of tumor vasculature, which, in turn, ledo significantly improved tumor control in melanoma andlioblastoma multiforme xenografts, which are otherwiseairly resistant to conventional doses of radiation (11).hese findings were further confirmed by Abdollahi et al.

44), who demonstrated that although VEGF and bFGF can

ncrease the radioresistance of endothelial cells in culture, c

U5416, a potential VEGFR RTK inhibitor when combinedith conventional doses (2 Gy) of radiation, led to reduced

ndothelial cell tubule formation and migration and signif-cantly decreased the surviving fraction of HUVECs inlonogenic assay (44). This same group also demonstratedhat triple combination of VEGFR inhibition with SU5416,hemotherapy (premetrexed), and radiation significantly in-reased the inhibition of proliferation, migration, tubuleormation, apoptosis, and clonogenic survival of endothelialells in culture. Interestingly, they demonstrated a link tonhibition of Akt phosphorylation by SU5416 as a potentialechanism of this effect (45). Avastin (Bevacizumab) is aonoclonal antibody against VEGF, a ligand for VEGFR,

pproved for treatment of metastatic colorectal cancer pa-ients when combined with chemotherapy (46). Phase Itudy of Avastin with concurrent radiotherapy and capecit-bine in locally advanced pancreatic cancer has recentlyeen reported, and final publications of these results areending (47).Broad-spectrum RTK inhibitors have the advantage of

argeting multiple RTKs that may be present in the tumor,ts microenvironment, or both. One such small molecule isTK787 (Novartis). PTK787 belongs to chemical class ofminophthalazines and is an oral inhibitor of the VEGFR2/DR/flk-1 tyrosine kinase (IC50 � 37 nM/L). It also exhib-

ts activity against flt-1, PDGFR-�, and c-Kit (IC50 � 770,80, and 730 nM/L, respectively) (48). It is not activegainst EGFR, FGFR, c-Met, Tie-2, c-Src, or c-Abl (48).TK787 in combination with radiation induced a dose-ependent reduction of proliferation of HUVECs but did notause these cells to undergo apoptosis. Treatment of p53-ysfunctional SW480 xenografts in vivo with PTK787 (4ays at 100 mg/kg/day) and radiation (4 days � 3 Gy) ledo increased tumor-growth delay compared with treatmentith either agent alone. Independently, each treatment mo-ality had minimal effect on tumor size and neovascular-zation. Significant reduction in microvessel density of theseumors was noted, which suggested that the combined ther-py was effectively targeting the tumor vasculature (49).imilarly, 2 broad-spectrum compounds from SUGENnow Pfizer), SU11248 and SU6668, have demonstratedignificant radiation sensitization effects in preclinical mod-ls. SU6668 was designed to inhibit kinase activity of FGF,DGF, and VEGF receptors (50, 51). Griffin et al. (52)emonstrated that combination of SU6668 with a singleigh dose (15 Gy) of radiation was significantly more ef-ective at controlling tumor growth compared with eitherodality alone. This observation was primarily attributed to

he increased radiosensitivity of tumor blood vessels. Fur-hermore, Lu et al. (53) demonstrated that concurrent ad-

inistration of SU6668 with fractionated conventionaloses of IR (3 Gy � 7 fractions) led to improved tumor-rowth delay compared with either treatment alone in LLCnd GL261 xenograft models. Akt phosphorylation inducedy IR was inhibited in endothelial cells that were pretreatedith SU6668 in vitro (53). Unfortunately, because of unac-

eptable toxicity profiles in clinical studies, SU6668 and

SsRibsnccbhttpgltsccioxdsDowomlddSltttst

tvtcVcf

Pt

tpvfs

liv

dtloLbterwiDH

Fstr1mSat(Fmi4

41Molecular strategies targeting the host component of cancer ● D. W. KIM et al.

U5416 are no longer being investigated in the clinicaletting (32). However, SU11248, another broadspectrumTK inhibitor from SUGEN/Pfizer, is still under active

nvestigation. SU11248 is an orally available indolinone-ased synthetic molecule, which was identified as a low-nMelective inhibitor of the angiogenic receptor tyrosine ki-ases Flk-1/KDR and PDGFR in both biochemical andellular assays (54). SU11248 was also found to inhibitellular signaling via Kit and FLT3. SU11248 exhibitedroad and potent antitumor activity in a wide range ofuman tumor xenografts in mice (A431 human epidermoidumors, Colo205 human colon tumors, H460 human lungumors, MDA-MB-435 human breast tumors, PC3 humanrostate tumors, SF763T human glioma) and in C6 ratlioma xenografts in mice (32, 54). Recent work from ouraboratory has demonstrated that SU11248 enhances radia-ion-induced endothelial cytotoxicity (13). Part of this re-ponse was the result of increased apoptosis in endothelialells treated with combination of SU11248 and RT. Thisondition was reflected by destruction of tumor vasculaturen a tumor vascular window model. We have further dem-nstrated that this tumor blood flow destruction in tumorenografts treated with conventional fractionated radiationoses (3 Gy � 5) and SU11248 can be assessed noninva-ively via imaging technology such as amplitude-modulatedoppler sonography (Fig. 2). Noninvasive imaging has thebvious advantage of being able to follow the same animalith serial measurements. For example, in Fig. 2, we dem-nstrate that by calculating power-weighted pixel density (aeasurement of vascularity), a marked reduction of vascu-

arity is noted in animals treated with SU11248 � RT for 5ays compared with either agent alone. We have furtheremonstrated that treatment of glioma xenografts withU11248 also leads to inhibition of PDGFR� phosphory-

ation, which suggests that the efficacy of this compound atumor control may also be the result of specific inhibition ofhe tumor cells that express PDGFR�. Furthermore, inhibi-ion of PDGFR� in the tumor cells may also cause increasedensitivity to the cytotoxic effects of IR directly in theseumors.

Thus, we have strong preclinical evidence that targetinghe upstream regulators of PI3K/Akt pathway in the tumorasculature is a potentially effective method of improvingumor therapy. This method is particularly attractive in theurrent clinical setting, as demonstrated by the numerousEGFR targeting agents being developed by the pharma-

eutical industry, which provide a potentially fertile area ofuture clinical trials that involve RT.

I3K as a direct target for radiation sensitization ofumor vasculature

Although upstream effectors such as RTKs are attractiveargets for therapy, the mechanism by which PI3K/Aktathway is activated in tumor vasculature exposed to con-entional doses of radiation is not fully understood. There-ore, other redundant pathways, such as VEGFR may pos-

ibly activate PI3K independent of RTKs. This prospect h

eads to the possibility that direct inhibition of PI3K activitys an even more efficacious method of sensitizing the tumorasculature to the cytotoxic effects of ionizing radiation.Targeted therapy by exploiting this pathway has been

emonstrated to increase radiation sensitivity of the endo-helial cells in culture and in xenograft models in vivo. Ouraboratory has demonstrated that inhibition of PI3K by usef extremely potent compounds such as wortmannin orY294002 (IC501.9 and 1.4 nM, respectively) when com-ined with conventional doses of IR led to effective destruc-ion of tumor vasculature in vivo and increased apoptosis ofndothelial cells in vitro (10). This process occurred even atelatively low concentrations of these compounds (4 nM forortmannin, and 2 �M for LY294002), which suggests that

t was likely through inhibition of PI3K, rather thanNA-PK or other potential targets of these compounds.owever, the potential effects that these compounds could

ig. 2. Tumor blood-flow analysis. Amplitude-modulated doppleronography was used to quantify microscopic blood flow in LLCumors implanted into the hind limb of C57BL6 mice. Shown areepresentative images of intensity of blood flow in tumors at Day0 of treatment. Tumors were imaged on Days 1 and 5 of treat-ent. Main indicates tumors that were treated with maintenanceU11248 alone without irradiation. The bar graphs represent theverage blood flow to the peripheral portion of the tumor graftsreated as described above. Bars indicate standard error. Asterisks*) indicate p � 0.05. PWPD � Power-weighted pixel density.rom Schuenemann AJ, Himmelfarb E, Geng L, et al. SU11248aintenance therapy prevents tumor regrowth after fractionated

rradiation of murine tumor models. Cancer Research 2003;63:009–4016, reproduced with permission.)

ave on other kinases, such as DNA-PK or CK2, were a

pihtfsLc

posascedPpctaconc

bamsiitIPllpumwvfcesti

Dt

AitN

cFrAdnpRedmPopTPil

R

wsmdwSfx(tnamtlcgsd

I

tseottct

42 I. J. Radiation Oncology ● Biology ● Physics Volume 64, Number 1, 2006

otential limitation of this study. These compounds can alsonhibit most isoforms of PI3K (18). Furthermore, studiesave indicated that radiation sensitization can also occur inumor cells when these compounds are used (55–58). There-ore, the lack of selectivity of these compounds, the lack oftability of wortmannin, and the lack of solubility ofY294002 are some reasons that have impeded furtherlinical studies of these agents.

More specific inhibition of this pathway, as a proof of theossibility, was achieved by use of adenovirus-mediatedverexpression of �85, a dominant negative mutant of p85ubunit of PI3K in endothelial cells (9). The transduction ofdenovirus (Ad) �85 leads to attenuation of the downstreamignaling of PI3K by preventing the recruitment of the p110omponent, which, thereby, inhibits PI3K function. Inter-stingly, Ad �85 transduction attenuated the radiation-in-uced phosphorylation of Akt. Furthermore, inhibition ofI3K in this manner led to increased endothelial cell apo-tosis when treated with 3 Gy of IR, with release of cyto-hrome C and caspase 3 and 9 cleaved products (9). Al-hough these studies demonstrated the importance ofctivation of PI3K/Akt activity in conferring endothelialell’s resistance to the cytotoxic effects of IR, the limitationf this study was that adenoviral delivery of dominantegative constructs are as of yet difficult to achieve in alinical setting.

Clinically relevant methods of inhibiting PI3K pathwayy use of pharmacological compounds are difficult tochieve because of the ubiquitous presence of PI3K in manyammalian cells. Such compounds, therefore, may render

ignificant toxicity. Most recently, ICOS Corporation hasntroduced a number of compounds that target specificsoforms of PI3K, in an attempt to confer improved selec-ivity of the compound for appropriate cellular targets.C486068 from ICOS targets the p110 � isoform of theI3K proteins. P110 � was found to be expressed in HL60

eukemia cells and in endothelial cells, which suggests someevel of cell-type specificity (12). Furthermore, this com-ound demonstrated no inhibitory effect of DNA-PK (59),nlike other nonspecific inhibitors of PI3K, such as wort-annin or LY294002. Most importantly, this compoundas highly effective at inhibiting PI3K/Akt pathway in

itro. Combination therapy of IC486068 with conventionalractionated doses of IR led to increased apoptosis, de-reased clonogenicity, and decreased migration of humanndothelial cells in vitro and led to tumor vasculature de-truction in vivo (12). This finding translated to increasedumor growth delay in xenograft models of GL261 and LLCn vivo (12).

ownstream target for radiation sensitization ofumor vasculature

A significant number of downstream effectors of PI3K/kt pathway have been identified and include pathways that

nvolve glycogen synthase kinase (GSK) 3�, mammalianarget of rapamycin (mTOR), FKHR, MDM2, BAD, and

F-�B (18). A number of these targets have molecular r

ompounds under preclinical and clinical investigations.or example, mTOR inhibitors under investigation includeapamycin and its derivatives (Rad001, CCI-779, andP23573) (60). Exposure of HUVECs to conventionaloses of IR has been demonstrated to activate mTOR sig-aling, presumably through the PI3K/Akt pathway (B. Lu,ersonal communication). Furthermore, mTOR inhibitorsAD001 and rapamycin were potent radiosensitizers ofndothelial cells in vitro and led to improved tumor-growthelay of glioma xenografts in vivo (B. Lu, personal com-unication). GSK-3� is another downstream target of theI3K/Akt pathway, and studies from our laboratory dem-nstrate that in endothelial cells, radiation leads to increasedhosphorylation of GSK-3�, which is mediated by PI3K.hese studies support the idea that downstream targets ofI3K/Akt pathway are relevant areas of investigation for

mproving the cytotoxic effects of radiation on the endothe-ium.

ole of adjuvant or maintenance therapyAlthough inhibition of PI3K/Akt pathway in combination

ith RT clearly leads to improved tumor control, we ob-erved that tumors can rapidly resume growth in test ani-als when combined therapy with SU11248 and IR are

iscontinued (13). Interestingly, persistent tumor controlas achieved by adjuvant/maintenance therapy withU11248 (13), compared with animals that did not receiveurther maintenance therapy. Similar results were seen inenograft models treated with antiangiogenic agents alone61). A potential advantage of maintenance therapy withhese agents is that resistance to this form of therapy doesot seem to develop, contrary to traditional chemotherapygents for which target cancer cells are prone to developultidrug resistance (61). Implications are that maintenance

herapy with antiangiogenic agents such as SU11248 mayead to reduced likelihood of early recurrence of cancer afteronclusion of definitive therapy. Furthermore, if tumor re-rowth is seen, salvage therapy by use of these agents maytill be an option, because the endothelium is unlikely toevelop resistance to these compounds.

NOVEL THERAPEUTIC STRATEGY:IDENTIFICATION OF NEOANTIGENS INDUCEDN THE TUMOR OR ITS MICROENVIRONMENT

IN RESPONSE TO COMBINED RTKINHIBITOR � RADIATION THERAPY

One of the most desirable attributes in targeted therapy ishe concept of specific targeting of tumors and relativeparing of normal tissues. In fact, many of these highlyffective compounds in preclinical studies fail to progressn to clinical studies because of the unacceptable normal-issue toxicity that can occur in patients. Although most ofhese toxicities are introduced early in the course of therapy,hronic adverse effects could also occur and, thereby, limithe potential use of these compounds. For example, the

ecent reports of increased cardiovascular toxicity of

Ct

alphrltab(rmd

Poaothrhpda

fstoplowtceqttpwbwwiscbRcanX

43Molecular strategies targeting the host component of cancer ● D. W. KIM et al.

OX-2 inhibitors will lead to questions about the future ofhis compound in treatment of patients with or without RT.

A novel strategy introduced initially by Arap, Pasqualini,nd Rouslahti (62, 63) and Pasqualini, Koivunen, and Rous-ahti (64) involves use of phage display libraries to isolateeptides that home specifically to tumor blood vessels. Theyave identified several peptides that target the �v integrineceptor (64), and coupling doxorubicin to these peptidesed to remarkably improved tumor control, with reducedoxicity in preclinical setting (63). We have used a similarpproach to successfully identify peptide sequences thatind to irradiated tumor vasculature in xenograft models65, 66). These results are the impetus for development of aadiation-guided drug delivery system to tumors and itsicroenvironment as a novel paradigm for targeted drug

elivery.We have demonstrated that combination therapy of

I3K/Akt inhibitors (PAIs) and a clinically relevant dosef radiation leads to significant disruption of the tumornd its microenvironment. We define PAIs as compoundsr drugs that lead to eventual downstream down-regula-ion of the PI3K/Akt pathway, including the RTKs weave described above, and, thereby, lead to increasedadiosensitivity. We make this distinction because weypothesize that the neoantigens produced by this ap-roach with any of the variety of PAIs (including RTKs,irect inhibitors of PI3K, or downstream effectors suchs rapamycin) and RT should have similar applicability

(A) RT (B) CONTROL

Fig. 3. Mice treated with SU11248�RT shows increasenude mice with D54 human glioblastoma cells implanteGy), (B) untreated, (C) 80 mg/kg SU11248 � RT (3 Gyafter therapy, the animals received intravenous injectioHVGGSV. Animals were imaged periodically (from 2 tocan detect the fluorescence emitted by the Cy-7 fluoroch24 hours. A representative image is shown. Tumors

Xenogen-IVIS as indicated (A–D).

or our targeted drug delivery approach. In our initialtudies, we have demonstrated increased apoptosis ofumor endothelial cells within 24 hours of a single dosef SU11248 � 3-Gy radiation (13). Therefore, we hy-othesized that this mode of therapy would have a highikelihood of induction of neoantigens within the tumorr its microenvironment. To begin to test this hypothesis,e selected peptides that bind preferentially to tumors

reated with PAI (in this case an RTK SU11248) �onventional-dose RT by use of a phage display libraryngineered to express 106 to 108 unique peptide se-uences on its capsid. Phages that bind to the treatedumors were recovered, and this process was repeatedhrough 6 passages to enrich for the specifically bindinghages. Phages bearing the peptide sequence HVGGSVere recovered from tumors treated with SU11248� RTut not from untreated control tumors. To directly testhether this peptide binds to tumors that are pretreatedith SU11248/RT combination therapy, we performed an

n vivo imaging experiment. Phages bearing the peptideequence HVGGSV were labeled with Cy-7 fluoro-hrome. These labeled phages were injected into miceearing tumor xenografts 6 hours after treatment withT, vehicle, SU11248�RT, or SU11248 alone as indi-ated (Fig. 3A–3D). The animals were then imaged undernesthesia by a Xenogen-IVIS System. Cy-7 emits atear-infrared wavelengths that can be visualized by theenogen-IVIS system. Preliminary studies indicate that

C) SU11248 + RT D) SU11248

ing of phage that express peptide HVGGSV. Athymictheir hindlimbs bilaterally were treated with (A) RT (3D) 80 mg/kg of SU11248 alone as indicated. Six hours10 to 1011 Cy-7–labeled phage that expressed peptideurs) on Xenogen-IVIS infrared imaging system, whichaximal binding of the Cy-7–labeled phage occurred at

the mice were then removed and re-imaged on the

d bindd into), or (

n of 1024 ho

rome. Mfrom

ttmaftiIStceptwaraosffcfb

obil(ii

beRtdtatpowiFo

1

44 I. J. Radiation Oncology ● Biology ● Physics Volume 64, Number 1, 2006

his peptide sequence binds with higher affinity to tumorsreated with SU11248�RT (Fig. 3C) compared with tu-ors that were treated with either agent alone (Fig. 3A

nd 3D) or untreated (Fig. 3B). Because of the potentialor artifactual fluorescence attenuation from the skin ofhe mice, we excised the tumors from the animals andmaged and quantitated them directly in the Xenogen-VIS system (Fig. 3A–3D). The tumors treated withU11248 � RT maintained a higher level of binding of

he Cy-7–labeled phage peptide HVGGSV, which furtheronfirmed our in vivo observation. On the basis of thisncouraging preliminary data, our laboratory is in therocess of identifying other peptide sequences that bindo the treated tumors, and, more importantly, we areorking on identification of inducible neoantigens that

re binding to these peptides. We are encouraged by ouresults, which demonstrate that even within the samenimal that received systemic SU11248, increased bindingccurs only in the presence of RT (Fig 3C). This resulttrongly suggests that the combination therapy is importantor this response. We do realize that potential confoundingactors exists, such as the reported abscopal effects, whichan occur in the contralateral untreated tumors (67). There-ore, we are pursuing our current studies with single-tumor–

earing animals to confirm our findings. This novel method w

REFEREN

0. Edwards E, Geng L, Tan J, et al. Phosphatidylinositol 3-ki-

1

1

1

1

1

1

1

1

1

f targeted drug delivery system can target cells that haveeen pretreated with minimal doses of radiation � RTKnhibitors. As an example, we linked cytotoxic agents toigands that bind to the neoantigens induced by PAI � RT68). The goal of this approach of guided delivery of drugss to reduce normal-tissue toxicity and increase bioavailabil-ty of the cytotoxic agents to the tumor.

CONCLUSION

Radiation therapy as a modality continues to evolve. Recentiologic discoveries have led to the concept that tumor micro-nvironment is an effective target for cancer therapy, includingT. However, the inherent resistance of the tumor vasculature

o the cytotoxic effects of RT at conventional fractionatedoses (2–3 Gy) needs to be overcome before this mode ofherapy can become a reality. PI3K/Akt pathway activation isn important factor that contributes to the relative radioresis-ance of the tumor vasculature. Timely development of com-ounds that inhibit this pathway, rapid preclinical assessmentf these compounds, and quick translation of these findings inell-designed clinical studies will lead to validation of target-

ng RT not only to the tumor but also to its vasculature.urthermore, innovative strategies to improve targeted deliveryf cytotoxic therapy to the tumor and its microenvironment

ill lead to improved therapeutic index.

CES

1. De Vita VT Jr, Hellman S, Rosenberg SA. Cancer. Principlesand practice of oncology. Vol 1. 6th ed. Philadelphia: Lippin-cott Williams & Wilkins; 2001.

2. Fuks Z, Vlodavsky I, Andreeff M, et al. Effects of extracel-lular matrix on the response of endothelial cells to radiation invitro. Eur J Cancer 1992;28A:725–731.

3. Garcia-Barros M, Paris F, Cordon-Cardo C, et al. Tumorresponse to radiotherapy regulated by endothelial cell apopto-sis. Science 2003;300:1155–1159.

4. Paris F, Fuks Z, Kang A, et al. Endothelial apoptosis as theprimary lesion initiating intestinal radiation damage in mice.Science 2001;293:293–297.

5. Wachsberger P, Burd R, Dicker AP. Tumor response to ion-izing radiation combined with antiangiogenesis or vasculartargeting agents: Exploring mechanisms of interaction. ClinCancer Res 2003;9:1957–1971.

6. Wachsberger P, Burd R, Dicker AP. Improving tumor re-sponse to radiotherapy by targeting angiogenesis signalingpathways. Hematol Oncol Clin North Am 2004;18:1039 –1057, viii.

7. Mauceri HJ, Hanna NN, Beckett MA, et al. Combined effectsof angiostatin and ionizing radiation in antitumour therapy.Nature 1998;394:287–291.

8. Gorski DH, Beckett MA, Jaskowiak NT, et al. Blockage of thevascular endothelial growth factor stress response increasesthe antitumor effects of ionizing radiation. Cancer Res 1999;59:3374–3378.

9. Tan J, Hallahan DE. Growth factor-independent activation ofprotein kinase B contributes to the inherent resistance ofvascular endothelium to radiation-induced apoptotic response.Cancer Res 2003;63:7663–7667.

nase/Akt signaling in the response of vascular endothelium toionizing radiation. Cancer Res 2002;62:4671–4677.

1. Geng L, Donnelly E, McMahon G, et al. Inhibition of vascularendothelial growth factor receptor signaling leads to reversalof tumor resistance to radiotherapy. Cancer Res 2001;61:2413–2419.

2. Geng L, Tan J, Himmelfarb E, et al. A specific antagonist ofthe p110delta catalytic component of phosphatidylinositol 3=-kinase, IC486068, enhances radiation-induced tumor vasculardestruction. Cancer Res 2004;64:4893–4899.

3. Schueneman AJ, Himmelfarb E, Geng L, et al. SU11248maintenance therapy prevents tumor regrowth after fraction-ated irradiation of murine tumor models. Cancer Res 2003;63:4009–4016.

4. Datta K, Bellacosa A, Chan TO, et al. Akt is a direct target ofthe phosphatidylinositol 3-kinase: Activation by growth fac-tors, v-src and v-Ha-ras, in Sf9 and mammalian cells. J BiolChem 1996;271:30835–30839.

5. Vanhaesebroeck B, Alessi DR. The PI3K-PDK1 connection:More than just a road to PKB. Biochem J 2000;346:561–576.

6. Alessi DR, James SR, Downes CP, et al. Characterization ofa 3-phosphoinositide-dependent protein kinase which phos-phorylates and activates protein kinase Balpha. Curr Biol1997;7:261–269.

7. Contessa JN, Hampton J, Lammering G, et al. Ionizing radi-ation activates Erb-B receptor dependent Akt and p70 S6kinase signaling in carcinoma cells. Oncogene 2002;21:4032–4041.

8. Stein RC. Prospects for phosphoinositide 3-kinase inhibi-tion as a cancer treatment. Endocr Relat Cancer 2001;8:237–248.

9. Hiles ID, Otsu M, Volinia S, et al. Phosphatidylinositol 3-ki-

2

2

2

2

2

2

2

2

2

2

3

3

3

3

3

3

3

3

3

3

4

4

4

4

4

4

4

4

4

4

5

5

5

5

5

5

5

5

45Molecular strategies targeting the host component of cancer ● D. W. KIM et al.

nase: Structure and expression of the 110 kd catalytic subunit.Cell 1992;70:419–429.

0. Klippel A, Escobedo JA, Hu Q, et al. A region of the 85-kilodalton (kDa) subunit of phosphatidylinositol 3-kinasebinds the 110-kDa catalytic subunit in vivo. Mol Cell Biol1993;13:5560–5566.

1. Klippel A, Escobedo JA, Hirano M, et al. The interaction ofsmall domains between the subunits of phosphatidylinositol3-kinase determines enzyme activity. Mol Cell Biol 1994;14:2675–2685.

2. Stern DF. Tyrosine kinase signalling in breast cancer: ErbBfamily receptor tyrosine kinases. Breast Cancer Res 2000;2:176–183.

3. Neve RM, Sutterluty H, Pullen N, et al. Effects of oncogenicErbB2 on G1 cell cycle regulators in breast tumour cells.Oncogene 2000;19:1647–1656.

4. Altiok N, Altiok S, Changeux JP. Heregulin-stimulated ace-tylcholine receptor gene expression in muscle: Requirementfor MAP kinase and evidence for a parallel inhibitory pathwayindependent of electrical activity. Embo J 1997;16:717–725.

5. Downward J. Lipid-regulated kinases: Some common themesat last. Science 1998;279:673–674.

6. Alessi DR, Andjelkovic M, Caudwell B, et al. Mechanism ofactivation of protein kinase B by insulin and IGF-1. Embo J1996;15:6541–6551.

7. Wymann MP, Pirola L. Structure and function of phosphoino-sitide 3-kinases. Biochim Biophys Acta 1998;1436:127–150.

8. Vivanco I, Sawyers CL. The phosphatidylinositol 3-kinase AKTpathway in human cancer. Nat Rev Cancer 2002;2:489–501.

9. Dudek H, Datta SR, Franke TF, et al. Regulation of neuronalsurvival by the serine-threonine protein kinase Akt. Science1997;275:661–665.

0. Datta SR, Dudek H, Tao X, et al. Akt phosphorylation of BADcouples survival signals to the cell-intrinsic death machinery.Cell 1997;91:231–241.

1. Zingg D, Riesterer O, Fabbro D, et al. Differential activationof the phosphatidylinositol 3=-kinase/Akt survival pathway byionizing radiation in tumor and primary endothelial cells.Cancer Res 2004;64:5398–5406.

2. Kim DW, Lu B, Hallahan DE. Receptor tyrosine kinase in-hibitors as anti-angiogenic agents. Curr Opin Investig Drugs2004;5:597–604.

3. Prewett M, Huber J, Li Y, et al. Antivascular endothelialgrowth factor receptor (fetal liver kinase 1) monoclonal anti-body inhibits tumor angiogenesis and growth of several mouseand human tumors. Cancer Res 1999;59:5209–5218.

4. Dunn IF, Heese O, Black PM. Growth factors in gliomaangiogenesis: FGFs, PDGF, EGF, and TGFs. J Neurooncol2000;50:121–137.

5. Fuks Z, Persaud RS, Alfieri A, et al. Basic fibroblast growthfactor protects endothelial cells against radiation-induced pro-grammed cell death in vitro and in vivo. Cancer Res 1994;54:2582–2590.

6. Seghezzi G, Patel S, Ren CJ, et al. Fibroblast growth factor-2(FGF-2) induces vascular endothelial growth factor (VEGF)expression in the endothelial cells of forming capillaries: Anautocrine mechanism contributing to angiogenesis. J Cell Biol1998;141:1659–1673.

7. Tsai JC, Goldman CK, Gillespie GY. Vascular endothelialgrowth factor in human glioma cell lines: Induced secretion byEGF, PDGF-BB, and bFGF. J Neurosurg 1995;82:864–873.

8. Witte L, Fuks Z, Haimovitz-Friedman A, et al. Effects ofirradiation on the release of growth factors from culturedbovine, porcine, and human endothelial cells. Cancer Res1989;49:5066–5072.

9. Schlessinger J. Cell signaling by receptor tyrosine kinases.

Cell 2000;103:211–225.

0. Claesson-Welsh L. Signal transduction by the PDGF recep-tors. Prog Growth Factor Res 1994;5:37–54.

1. Claesson-Welsh L. Platelet-derived growth factor receptorsignals. J Biol Chem 1994;269:32023–32026.

2. Kim DW, Choy H. Potential role for epidermal growth factorreceptor inhibitors in combined-modality therapy for non-small-cell lung cancer. Int J Radiat Oncol Biol Phys 2004;59:11–20.

3. Kozin SV, Boucher Y, Hicklin DJ, et al. Vascular endothelialgrowth factor receptor-2-blocking antibody potentiates radia-tion-induced long-term control of human tumor xenografts.Cancer Res 2001;61:39–44.

4. Abdollahi A, Lipson KE, Han X, et al. SU5416 and SU6668attenuate the angiogenic effects of radiation-induced tumorcell growth factor production and amplify the direct anti-endothelial action of radiation in vitro. Cancer Res 2003;63:3755–3763.

5. Bischof M, Abdollahi A, Gong P, et al. Triple combination ofirradiation, chemotherapy (pemetrexed), and VEGFR inhibi-tion (SU5416) in human endothelial and tumor cells. Int JRadiat Oncol Biol Phys 2004;60:1220–1232.

6. Hurwitz H, Fehrenbacher L, Novotny W, et al. Bevacizumabplus irinotecan, fluorouracil, and leucovorin for metastaticcolorectal cancer. N Engl J Med 2004;350:2335–2342.

7. Crane CH, Ellis LM, O’Reilly MS, et al. RhuMab VEGF(Bevacizumab) with concurrent radiotherapy and capecitabinein locally advanced pancreatic cancer: An active, well toler-ated regimen. Int J Radiat Oncol Biol Phys 2004;60(Suppl.1):S149.

8. Wood JM, Bold G, Buchdunger E, et al. PTK787/ZK 222584,a novel and potent inhibitor of vascular endothelial growthfactor receptor tyrosine kinases, impairs vascular endothelialgrowth factor-induced responses and tumor growth after oraladministration. Cancer Res 2000;60:2178–2189.

9. Hess C, Vuong V, Hegyi I, et al. Effect of VEGF receptorinhibitor PTK787/ZK222584 combined with ionizing radia-tion on endothelial cells and tumour growth. Br J Cancer2001;85:2010–2016.

0. Laird AD, Christensen JG, Li G, et al. SU6668 inhibits Flk-1/KDR and PDGFRbeta in vivo, resulting in rapid apoptosis oftumor vasculature and tumor regression in mice. Faseb J2002;16:681–690.

1. Laird AD, Vajkoczy P, Shawver LK, et al. SU6668 is a potentantiangiogenic and antitumor agent that induces regression ofestablished tumors. Cancer Res 2000;60:4152–4160.

2. Griffin RJ, Williams BW, Wild R, et al. Simultaneous inhi-bition of the receptor kinase activity of vascular endothelial,fibroblast, and platelet-derived growth factors suppresses tu-mor growth and enhances tumor radiation response. CancerRes 2002;62:1702–1706.

3. Lu B, Geng L, Musiek A, et al. Broad spectrum receptortyrosine kinase inhibitor, SU6668, sensitizes radiation viatargeting survival pathway of vascular endothelium. Int JRadiat Oncol Biol Phys 2004;58:844–850.

4. Mendel DB, Laird AD, Xin X, et al. In vivo antitumor activityof SU11248, a novel tyrosine kinase inhibitor targeting vas-cular endothelial growth factor and platelet-derived growthfactor receptors: Determination of a pharmacokinetic/pharma-codynamic relationship. Clin Cancer Res 2003;9:327–337.

5. Gupta AK, Cerniglia GJ, Mick R, et al. Radiation sensitizationof human cancer cells in vivo by inhibiting the activity of PI3Kusing LY294002. Int J Radiat Oncol Biol Phys 2003;56:846–853.

6. Sarkaria JN, Tibbetts RS, Busby EC, et al. Inhibition ofphosphoinositide 3-kinase related kinases by the radiosensi-tizing agent wortmannin. Cancer Res 1998;58:4375–4382.

7. Rosenzweig KE, Youmell MB, Palayoor ST, et al. Radiosen-

sitization of human tumor cells by the phosphatidylinositol3-

5

5

6

6

6

6

6

6

6

6

6

46 I. J. Radiation Oncology ● Biology ● Physics Volume 64, Number 1, 2006

kinase inhibitors wortmannin and LY294002 correlates withinhibition of DNA-dependent protein kinase and prolongedG2-M delay. Clin Cancer Res 1997;3:1149–1156.

8. Price BD, Youmell MB. The phosphatidylinositol 3-kinaseinhibitor wortmannin sensitizes murine fibroblasts and humantumor cells to radiation and blocks induction of p53 followingDNA damage. Cancer Res 1996;56:246–250.

9. Sadhu C, Dick K, Treiberg J. Inhibitors of human phos-phatidylinositol 3-kinase delta. US Patent Application20020161014 2002;WO 01/81346.

0. Chan S. Targeting the mammalian target of rapamycin(mTOR): A new approach to treating cancer. Br J Cancer2004;91:1420–1424.

1. Boehm T, Folkman J, Browder T, et al. Antiangiogenic ther-apy of experimental cancer does not induce acquired drugresistance. Nature 1997;390:404–407.

2. Arap W, Pasqualini R, Ruoslahti E. Chemotherapy targeted to

tumor vasculature. Curr Opin Oncol 1998;10:560–565.

3. Arap W, Pasqualini R, Ruoslahti E. Cancer treatment bytargeted drug delivery to tumor vasculature in a mouse model.Science 1998;279:377–380.

4. Pasqualini R, Koivunen E, Ruoslahti E. Alpha v integrins asreceptors for tumor targeting by circulating ligands. Nat Bio-technol 1997;15:542–546.

5. Hallahan D, Geng L, Qu S, et al. Integrin-mediated targetingof drug delivery to irradiated tumor blood vessels. Cancer Cell2003;3:63–74.

6. Hallahan DE, Geng L, Cmelak AJ, et al. Targeting drugdelivery to radiation-induced neoantigens in tumor microvas-culature. J Control Release 2001;74:183–191.

7. Camphausen K, Moses MA, Menard C, et al. Radiation ab-scopal antitumor effect is mediated through p53. Cancer Res2003;63:1990–1993.

8. Geng L, Osusky K, Konjeti S, et al. Radiation-guided drugdelivery to tumor blood vessels results in improved tumor

growth delay. J Control Release 2004;99:369–381.