Trimodal Cancer Treatment: Beneficial Effects of Combined ... · Trimodal Cancer Treatment:...

14
Trimodal Cancer Treatment: Beneficial Effects of Combined Antiangiogenesis, Radiation, and Chemotherapy Peter E. Huber, 1 Marc Bischof, 1 Ju ¨rgen Jenne, 1 Sabine Heiland, 3 Peter Peschke, 1 Rainer Saffrich, 1 Hermann-Josef Gro ¨ne, 2 Ju ¨rgen Debus, 1 Kenneth E. Lipson, 4 and Amir Abdollahi 1 Departments of 1 Radiation Oncology and 2 Molecular Pathology, German Cancer Research Center and 3 Department of Neuroradiology, University of Heidelberg Medical School, Heidelberg, Germany; and 4 SUGEN, Inc., South San Francisco, California Abstract It has been suggested that chemotherapy and radiotherapy could favorably be combined with antiangiogenesis in dual anticancer strategy combinations. Here we investigate the effects of a trimodal strategy consisting of all three therapy approaches administered concurrently. We found that in vitro and in vivo , the antiendothelial and antitumor effects of the triple therapy combination consisting of SU11657 (a multi- targeted small molecule inhibitor of vascular endothelial growth factor and platelet-derived growth factor receptor tyrosine kinases), Pemetrexed (a multitargeted folate anti- metabolite), and ionizing radiation were superior to all single and dual combinations. The superior effects in human umbilical vein endothelial cells and tumor cells (A431) were evident in cell proliferation, migration, tube formation, clonogenic survival, and apoptosis assays (sub-G 1 and caspase-3 assessment). Exploring potential effects on cell survival signaling, we found that radiation and chemother- apy induced endothelial cell Akt phosphorylation, but SU11657 could attenuate this process in vitro and in vivo in A431 human tumor xenografts growing s.c. on BALB/c nu /nu mice. Triple therapy further decreased tumor cell prolifera- tion (Ki-67 index) and vessel count (CD31 staining), and induced greater tumor growth delay versus all other therapy regimens without increasing apparent toxicity. When testing different treatment schedules for the A431 tumor, we found that the regimen with radiotherapy (7.5 Gy single dose), given after the institution of SU11657 treatment, was more effective than radiotherapy preceding SU11657 treatment. Accordingly, we found that SU11657 markedly reduced intratumoral interstitial fluid pressure from 8.8 F 2.6 to 4.2 F 1.5 mm Hg after 1 day. Likewise, quantitative T2- weighed magnetic resonance imaging measurements showed that SU11657-treated mice had reduced intratumoral edema. Our data indicates that inhibition of Akt signaling by antiangiogenic treatment with SU11657 may result in: ( a ) normalization of tumor blood vessels that cause prerequisite physiologic conditions for subsequent radio/chemotherapy, and (b) direct resensitization of endothelial cells to radio/ chemotherapy. We conclude that trimodal cancer therapy combining antiangiogenesis, chemotherapy, and radiotherapy has beneficial molecular and physiologic effects to emerge as a clinically relevant antitumor strategy. (Cancer Res 2005; 65(9): 3643-55) Introduction The concurrent or sequential combination of radiotherapy and chemotherapy is now considered a standard therapy regimen for the treatment of many tumors (1–4). The underlying rationale for combining different treatment modalities is to broaden the therapeutic index. It is assumed that different treatment modalities have overlapping anticancer effects but a decreased overlapping spectrum of side effects. Antiangiogenesis is one of the most promising new therapy principles which has recently gained strong impetus from successful clinical studies with an anti–vascular endothelial growth factor (VEGF) antibody (4–7). It has been suggested in experimental and clinical studies that angiogenesis inhibitors may be favorably combined with either chemotherapy (1, 4, 5, 8) or radiotherapy in dual combinations (3, 9–17). Therefore, the idea pursued in this paper of investigating a trimodal therapy regimen seems logical. The rationale for combining radiation with angiogenesis inhibitors is derived, in part, from findings that irradiation induces expression of proangiogenic cytokines such as VEGF or platelet-derived growth factor (PDGF), resulting in protection of vessels from radiation-induced cell damage (9, 11, 18). Clinically, elevated expression of these growth factors correlates with higher vessel density and negative prognosis in various tumors (4, 10). Moreover, such tumors are often relatively resistant to radiation therapy (4, 10). In addition to promoting the expression of proangiogenic cytokines, radiation has also been reported to kill endothelial cells (9). Thus, the rationale for this combination involves both killing the endothelial cells with radiation and preventing their regrowth with the angiogenesis inhibitor. Similarly, the favorable combination of chemotherapeutic agents and angiogenesis inhibitors may be due to the sustained antiangiogenic effects of the chemotherapeutic agents (3, 4, 19). Therefore, the question arises of whether the combination of all three modalities would have beneficial cellular or physiologic effects that could provide a rationale for trimodal therapy. The purpose of the present study was to evaluate the molecular and physiologic effects of a trimodal anticancer regimen consisting of the angiogenesis inhibitor SU11657, the chemotherapeutic agent Pemetrexed, and ionizing radiation, on human endothelium and A431 human epidermoid cancer cells in vitro and A431 human tumor xenografts on BALB/c mice in vivo . SU11657 is a multitargeted inhibitor of class III/V receptor tyrosine kinases with potency and selectivity similar to SU11248, Note: P.E. Huber and M. Bischof share first authorship. Requests for reprints: Peter E. Huber, Department of Radiation Oncology, German Cancer Research Center, 280 Im Neuenheimer Feld, Heidelberg 69120, Germany. Phone: 49-6221-42-2515; Fax: 49-6221-42-2514; E-mail: [email protected]. I2005 American Association for Cancer Research. www.aacrjournals.org 3643 Cancer Res 2005; 65: (9). May 1, 2005 Research Article Research. on February 25, 2021. © 2005 American Association for Cancer cancerres.aacrjournals.org Downloaded from

Transcript of Trimodal Cancer Treatment: Beneficial Effects of Combined ... · Trimodal Cancer Treatment:...

Page 1: Trimodal Cancer Treatment: Beneficial Effects of Combined ... · Trimodal Cancer Treatment: Beneficial Effects of Combined Antiangiogenesis, Radiation, and Chemotherapy Peter E. Huber,

Trimodal Cancer Treatment: Beneficial Effects of Combined

Antiangiogenesis, Radiation, and Chemotherapy

Peter E. Huber,1Marc Bischof,

1Jurgen Jenne,

1Sabine Heiland,

3Peter Peschke,

1

Rainer Saffrich,1Hermann-Josef Grone,

2Jurgen Debus,

1

Kenneth E. Lipson,4and Amir Abdollahi

1

Departments of 1Radiation Oncology and 2Molecular Pathology, German Cancer Research Center and3Department of Neuroradiology, University of Heidelberg Medical School, Heidelberg, Germany;and 4SUGEN, Inc., South San Francisco, California

Abstract

It has been suggested that chemotherapy and radiotherapycould favorably be combined with antiangiogenesis in dualanticancer strategy combinations. Here we investigate theeffects of a trimodal strategy consisting of all three therapyapproaches administered concurrently. We found that in vitroand in vivo , the antiendothelial and antitumor effects of thetriple therapy combination consisting of SU11657 (a multi-targeted small molecule inhibitor of vascular endothelialgrowth factor and platelet-derived growth factor receptortyrosine kinases), Pemetrexed (a multitargeted folate anti-metabolite), and ionizing radiation were superior to allsingle and dual combinations. The superior effects in humanumbilical vein endothelial cells and tumor cells (A431) wereevident in cell proliferation, migration, tube formation,clonogenic survival, and apoptosis assays (sub-G1 andcaspase-3 assessment). Exploring potential effects on cellsurvival signaling, we found that radiation and chemother-apy induced endothelial cell Akt phosphorylation, butSU11657 could attenuate this process in vitro and in vivo inA431 human tumor xenografts growing s.c. on BALB/c nu/numice. Triple therapy further decreased tumor cell prolifera-tion (Ki-67 index) and vessel count (CD31 staining), andinduced greater tumor growth delay versus all other therapyregimens without increasing apparent toxicity. When testingdifferent treatment schedules for the A431 tumor, we foundthat the regimen with radiotherapy (7.5 Gy single dose),given after the institution of SU11657 treatment, was moreeffective than radiotherapy preceding SU11657 treatment.Accordingly, we found that SU11657 markedly reducedintratumoral interstitial fluid pressure from 8.8 F 2.6 to4.2 F 1.5 mm Hg after 1 day. Likewise, quantitative T2-weighed magnetic resonance imaging measurements showedthat SU11657-treated mice had reduced intratumoral edema.Our data indicates that inhibition of Akt signaling byantiangiogenic treatment with SU11657 may result in: (a)normalization of tumor blood vessels that cause prerequisitephysiologic conditions for subsequent radio/chemotherapy,and (b) direct resensitization of endothelial cells to radio/chemotherapy. We conclude that trimodal cancer therapycombining antiangiogenesis, chemotherapy, and radiotherapy

has beneficial molecular and physiologic effects to emerge asa clinically relevant antitumor strategy. (Cancer Res 2005;65(9): 3643-55)

Introduction

The concurrent or sequential combination of radiotherapy andchemotherapy is now considered a standard therapy regimen forthe treatment of many tumors (1–4). The underlying rationalefor combining different treatment modalities is to broaden thetherapeutic index. It is assumed that different treatmentmodalities have overlapping anticancer effects but a decreasedoverlapping spectrum of side effects. Antiangiogenesis is one ofthe most promising new therapy principles which has recentlygained strong impetus from successful clinical studies with ananti–vascular endothelial growth factor (VEGF) antibody (4–7). Ithas been suggested in experimental and clinical studies thatangiogenesis inhibitors may be favorably combined with eitherchemotherapy (1, 4, 5, 8) or radiotherapy in dual combinations(3, 9–17). Therefore, the idea pursued in this paper ofinvestigating a trimodal therapy regimen seems logical.The rationale for combining radiation with angiogenesis

inhibitors is derived, in part, from findings that irradiationinduces expression of proangiogenic cytokines such as VEGF orplatelet-derived growth factor (PDGF), resulting in protection ofvessels from radiation-induced cell damage (9, 11, 18). Clinically,elevated expression of these growth factors correlates with highervessel density and negative prognosis in various tumors (4, 10).Moreover, such tumors are often relatively resistant to radiationtherapy (4, 10). In addition to promoting the expression ofproangiogenic cytokines, radiation has also been reported to killendothelial cells (9). Thus, the rationale for this combinationinvolves both killing the endothelial cells with radiation andpreventing their regrowth with the angiogenesis inhibitor.Similarly, the favorable combination of chemotherapeutic agentsand angiogenesis inhibitors may be due to the sustainedantiangiogenic effects of the chemotherapeutic agents (3, 4, 19).Therefore, the question arises of whether the combination of

all three modalities would have beneficial cellular or physiologiceffects that could provide a rationale for trimodal therapy. Thepurpose of the present study was to evaluate the molecular andphysiologic effects of a trimodal anticancer regimen consisting ofthe angiogenesis inhibitor SU11657, the chemotherapeutic agentPemetrexed, and ionizing radiation, on human endothelium andA431 human epidermoid cancer cells in vitro and A431 humantumor xenografts on BALB/c mice in vivo .SU11657 is a multitargeted inhibitor of class III/V receptor

tyrosine kinases with potency and selectivity similar to SU11248,

Note: P.E. Huber and M. Bischof share first authorship.Requests for reprints: Peter E. Huber, Department of Radiation Oncology, German

Cancer Research Center, 280 Im Neuenheimer Feld, Heidelberg 69120, Germany.Phone: 49-6221-42-2515; Fax: 49-6221-42-2514; E-mail: [email protected].

I2005 American Association for Cancer Research.

www.aacrjournals.org 3643 Cancer Res 2005; 65: (9). May 1, 2005

Research Article

Research. on February 25, 2021. © 2005 American Association for Cancercancerres.aacrjournals.org Downloaded from

Page 2: Trimodal Cancer Treatment: Beneficial Effects of Combined ... · Trimodal Cancer Treatment: Beneficial Effects of Combined Antiangiogenesis, Radiation, and Chemotherapy Peter E. Huber,

which has been studied in preclinical animal models (20–25).SU11248 has also shown promise in its phase I/II trials in patients(20–26). Both SU11248 and SU11657 exert their antiangiogeniceffects via potent inhibition of VEGF receptor 1 (VEGFR1),VEGFR2, and PDGF receptor (PDGFR; refs. 21, 22, 24). Additionally,both compounds may have direct antitumor effects by inhibition ofc-kit and fetal liver tyrosine kinase 3 ( flt3) expressed on tumor cells(21, 23, 25).In contrast with the widely used classic antifolates such as 5-

fluorouracil, the novel folate antimetabolite Pemetrexed inhibitsseveral key enzymes of thymidylate and purine synthesis, likethymidylate synthase, dihydrofolate reductase, and glycinamideribonucleotide formyl transferase, as well as of other folate-requiring enzymes (27). Pemetrexed exhibits significant antitumoractivity in a broad spectrum of human tumors, includingmesothelioma, pancreatic, colorectal, gastrointestinal, lung, headand neck, breast and cervix cancers, and has currently enteredphase III clinical trials (28). Antifolates have been established incancer treatment for many years and are widely used incombination with radiotherapy (29). The ability of Pemetrexed tosensitize cells to ionizing radiation was reported in preclinicalstudies (30–32).In this study, we have examined several variables, in vitro and

in vivo , as a function of therapy with different combinations ofSU11657, Pemetrexed, and radiation. For example, apoptosis hasbeen suggested to influence tumor response of radiotherapy,chemotherapy and antiangiogenesis. Evading apoptosis has beenshown to promote drug resistance (33) and survival signaling byAkt (also known as protein kinase B; refs. 34, 35). Furthermore,it has been shown that radiation induces the phosphorylation ofAkt via phosphatidylinositol 3V-kinase signaling (36, 37). Thus,the prosurvival effect of Akt activation in endothelial cellsprovides a key escape mechanism against radiation damage(36, 37). We therefore investigated treatment-related apoptosisinduction and the activation of Akt in endothelial cells. To testthe antiangiogenic effects of the treatments, we also didfunctional angiogenesis assays in human endothelial cells. InA431 tumor xenografts growing s.c. on nude mice, we examinedtumor growth and immunohistopathologic changes. In additionto cellular sensitivity, we were interested in the altered tumorphysiology associated with combinations of SU11657 and radio/chemotherapy. To investigate the physiologic effects, wemeasured intratumoral interstitial fluid pressure (IFP). It isassumed that elevated IFPs, a hallmark of solid tumors, may beassociated with reduced tumor blood flow and impaired deliveryof therapeutic drugs (3, 38–41) and should thus influence theresults of our combination therapy. The intratumoral IFP datawere correlated with functional T2-weighed magnetic resonanceimaging (MRI) scans which depict tumor morphology as well astumor physiologic variables such as intratumoral edema. Ourresults integrate cellular and physiologic effects of trimodaltherapy, and argue for the beneficial combined effects of atrimodal anticancer strategy.

Materials and Methods

Cell culture, drug exposure, and irradiation. Human epidermoid

carcinoma cells (A431) were obtained from the Tumorbank of theGerman Cancer Research Center (DKFZ, Heidelberg) and were grown as

monolayers in DMEM medium with 10% FCS. Primary isolated human

umbilical vein endothelial cells (HUVEC, Promocell, Heidelberg, Germany)

were cultured up to passage 6, maintained in culture at 37jC with 5%

CO2 and 95% humidity in serum-reduced (5% FCS) modified Promocellmedium supplemented with 2 ng/mL VEGF and 4 ng/mL basic fibroblast

growth factor (Promocell; refs. 9, 42). Pemetrexed was obtained from Lilly

Research Laboratories (Indianapolis, IN) and was dissolved in an aqueous

buffer [150 mmol/L phosphate (pH 7.4)]. Cells were treated withPemetrexed at various concentrations for 2 hours followed by a medium

change to remove the drug. SU11657 was provided by SUGEN, Inc.

(South San Francisco, CA). Cells were treated with SU11657 for 2 hours

in growth factor–reduced modified Promocell medium. Then the samevolume of modified Promocell medium with 2-fold standard growth

factor concentrations was added and cells were incubated under

standard conditions (9, 42). If not indicated otherwise, for combination

experiments, we chose 0.1 Amol/L SU11657 for proliferation andclonogenic survival assays, and 1 Amol/L SU11657 for other assays

involving Matrigel. These doses were determined in pilot experiments as

small doses showing significant effects as monotherapy. Photonirradiations from 0 to 8 Gy were done immediately after antiangiogenic

and/or chemotherapy treatment using a linear accelerator at 6 MV

(Mevatron, Siemens, Erlangen, Germany) at a dose rate of 2.5 Gy/minute.

Cell proliferation and clonogenic assay. Cell proliferation/cell

viability was measured as described previously (9, 42). Briefly, a

suspension of 50,000 endothelial or tumor cells was seeded (collagen

I-coated flasks for endothelial cells) and incubated for 24 hours under

standard conditions. The cells were then treated as indicated and

counted after another 72 hours of incubation. To measure the clonogenic

survival, endothelial or tumor cells were plated in triplicate to yield 50 to

100 colonies per culture flask and were incubated for 14 to 21 days.

Colonies of more than 50 cells, as assessed by microscopic inspection,

were scored as survivors, as described earlier (9, 31). For graphical

representation of the combination experiments, the mean values of the

measured surviving fractions were multiplied with the averaged surviving

fraction after drug exposure alone. Radiation doses that reduced cell

proliferation to 60% (D60 in Gy) following exposure to X-rays alone and

in the combination treatments were derived by linear interpolation

between measured data pairs. Enhancement ratios were calculated as the

quotient of D60 with irradiation alone and D60 of the respective dual or

triple combination. Sensitivity variables for clonogenic survival were

calculated accordingly, but using radiation doses that decreased survival

to 5% (D5).

Endothelial cell migration and tube formation. The endothelial cell

ability to form tubular structures was assessed as previously described(9). Briefly, 24-well plates were coated with Matrigel (Becton Dickinson,

Heidelberg, Germany). Endothelial cells (48,000 cells per well) undergo

differentiation into capillary-like tube structures when plated on Matrigel.

Cell were incubated with Pemetrexed (1.06 Amol/L) and/or SU11657(1 Amol/L). Then samples were irradiated with a single dose of 4 Gy, and

incubated for 6 hours on the Matrigel at 37jC/5% CO2. After 6 hours of

incubation, the media was aspirated, the cells were fixed and stained

with Diff-Quik II reagents (Dade Behring AG, Germany). Tubes werequantified by counting the number of anastomoses per field on at least

five wells per group.

The migration of HUVEC after exposure to different treatment regimenswas tested in a migration assay as described previously, with minor

modifications (9). Briefly, Matrigel-coated transwell inserts (8 Amol/L pore

size; Becton Dickinson) were used. Cells were incubated with Pemetrexed

(1.06 Amol/L) and/or SU11657 (1.0 Amol/L) for 2 hours and respectivesamples were irradiated with a single dose of 4 Gy. Then a cell suspension of

200 AL (3 � 105 cells/mL) per condition was added in triplicate transwells.

After 18 hours of incubation, endothelial cells that had invaded the

underside of the membrane were fixed, stained in thiazine and eosinesolution by using Diff-Quick II solution (Dade Behring) and sealed on slides.

Migrated cells were counted by microscopy.

Apoptosis in endothelial cells. HUVEC were treated with singlemodalities (0, 2, or 5 Gy irradiation; 1.06 Amol/L Pemetrexed; 1.0 Amol/L

SU11657) or combinations. 24 hours after the end of therapy, cells were

harvested and prepared for analysis of sub-G1 DNA by flow cytometry

(FACScan, Becton Dickinson) with propidium iodide staining (9, 43).

Cancer Research

Cancer Res 2005; 65: (9). May 1, 2005 3644 www.aacrjournals.org

Research. on February 25, 2021. © 2005 American Association for Cancercancerres.aacrjournals.org Downloaded from

Page 3: Trimodal Cancer Treatment: Beneficial Effects of Combined ... · Trimodal Cancer Treatment: Beneficial Effects of Combined Antiangiogenesis, Radiation, and Chemotherapy Peter E. Huber,

Histogram analysis (Modfit, Verity) of cells with sub-G1 DNA content wasdone after setting an analysis gate in the forward- versus sideward-

scattergram encompassing the major live cell population. To measure

caspase-3 activity (44), HUVEC were left untreated, exposed to single

modalities (irradiation, 4 Gy; Pemetrexed, 1.06 Amol/L; or SU11657,1.0 Amol/L) or respective combinations with and without irradiation or

were subjected to a triple-combination treatment. At 6 hours after

incubation, cells were washed twice with cold PBS, resuspended using the

Cytofix/Cytoperm solution (BD PharMingen), and incubated for 20 minuteson ice. Cells were pelleted, washed with washing buffer, and resuspended in

washing buffer plus PE-conjugated monoclonal active caspase-3

(BD PharMingen, catalogue No. 68655X) using 20 AL/1 � 106 cells and

incubated for 30 minutes at room temperature. Following incubation withthe antibody, cells were washed in washing buffer and resuspended in PBS

and analyzed by flow cytometry.

Immunocytochemistry. To analyze Akt phosphorylation, HUVEC were

exposed to single treatment modalities (irradiation, 0, 2, or 5 Gy;

Pemetrexed, 1.06 Amol/L; SU11657, 1.0 Amol/L) or combinations. Cells

were grown on glass coverslips and fixed 45 minutes after treatment in 3.7%

paraformaldehyde. Cells were incubated with a rabbit anti-Akt, phospho-

specific (Ser473) primary antibody (Santa Cruz, Heidelberg, Germany)

followed by incubation with the Alexa-488 conjugated anti-mouse

secondary antibody (Molecular Probes, Leiden, the Netherlands). Cells were

counterstained with propidium iodide for nuclear staining. Finally, the cells

were washed and mounted with Mowiol on microscope slides, observed on

a Zeiss Axiovert 10 inverted microscope with a 20� objective. Images were

acquired using a cooled CCD camera (Photometrics, CH250) using

fluorescence excitation with an FITC filter set and an acquisition time of

5 seconds, and then stored as TIFF files on a Sun SparcStation 20 Unix

workstation. Image processing and analysis was done with programs

written for the Khoros Software package. Averaged intensity F SD was

analyzed for 10 fields on five slides.

Animal studies. All in vivo experiments were approved by institutional

and governmental animal protection committees (Regierungsprasidium,

Karlsruhe, Germany). Athymic female mice (BALB/c, nu/nu , 8 weeks, 20 g)

were purchased from Charles River Laboratories (Sulzfeld, Germany).

Animals were maintained under clean room conditions in sterile rodent

microisolator cages (VentiRack, Heidelberg, Germany). Human A431

epidermoid carcinoma cells were injected s.c. into the right hind limb

(1 � 107 cells in 100 AL PBS). Two sets of experiments were done. The

first experiment set out to ask the question whether the order of

administration of single dose radiotherapy and continuous angiogenesis

inhibition influenced tumor growth. Mice with established tumors

[f200 mm3 (222 F 39 mm3) mean F SD] were stratified into five

groups (n = 10-12 each) receiving either vehicle as control, radiotherapy

alone, SU11657 alone, or two regimens of combined radiotherapy and

SU11657. Radiotherapy was given either 1 day before start of

antiangiogenic therapy or 1 day after start of antiangiogenic therapy, as

indicated.For the second set of experiments, three modalities, antiangiogenesis,

chemotherapy, and radiotherapy were combined in a trimodal schedule, as

indicated. Animals were randomized into nine groups (n = 12-15 each)

when tumor volume reached f200 mm3 (208 F 42 mm3, mean F SD).Mean tumor volume was determined thrice weekly by calipers and

calculated by volume V = length � width � width � 0.5.

Chemotherapy with Pemetrexed was administered in the trimodalexperiment at 150 mg/kg (total dose, 600 mg) in 200 AL PBS i.p. for

4 consecutive days (days 0-3), which we defined as a conventional schedule.

Additionally, a metronomic application of Pemetrexed (Pemetrexedmetron)

with a lower daily dosage (100 mg/kg) was given on days 0, 1, and 2, and ondays 7, 8, and 9 (same total dose of 600 mg).

SU11657 was given s.c. in 100 AL carboxymethylcellulose as vehicle thrice

weekly at 100 mg/kg starting on day 0 or day 1, as indicated, and was given

until the end of observation. In combination regimen, SU11657 was given30 minutes before Pemetrexed injection. Radiotherapy was delivered using a

Co-60 source (Gammatron, Siemens) with a single dose of 7.5 Gy. When

given on day 0, radiotherapy was given 24 hours before antiangiogenic

therapy. In the trimodal experiment, radiotherapy was given on day 1,4 hours after SU11657 and Pemetrexed administration.

Measurement of interstitial fluid pressure in vivo . IFP in A431

tumors was measured to determine the influence of the different therapy

modalities and its association with tumor growth and therapy outcome.

The IFP is thought to be nearly uniform throughout the central volume

of an idealized spherical tumor (12, 19). Here, the intratumoral IFP was

measured using a modified pressure sensor (SAMBA 3000, Samba

Sensors, Gothenburg, Sweden) consisting of a laser-optic fiber. The

pressure measurement is not based on the often used principle of a

hydrostatic difference between a glass capillary filled with fluids and the

interstitium, but is instead based on a pressure-sensitive optical Fabry-

Perot interferometer. According to the manufacturer, the fiber (diameter,

0.25 mm inside a 0.3 mm cannula) was calibrated between 0.95 and 1.15

bar with a resolution of 0.2 mbar (F2% accuracy). Thus, we measured

the IFP relative to the ambient atmospheric pressure, as assessed with

the built-in barometer of the SAMBA 3000. When inserted in the tumor

tissue, the pressure signal typically rose up to a plateau within

30 seconds. This equilibrium pressure at 1 minute was considered the

IFP. In prior experiments, we found that the IFP was independent of the

specific localization of the pressure sensor inside the tumors, thus

confirming the uniformity of the pressure distribution in this experi-

mental tumor system (data not shown).

Functional magnetic resonance imaging. MRI is a noninvasive

method to visualize tumor size and morphology. Two randomly chosenanimals from each therapy group were examined on day 10. The animals

were examined in a 2.35 T scanner (Biospec 24/40, Bruker Medizintechnik,

Ettlingen, Germany; ref. 45). An actively shielded gradient coil with a

120-cm inner diameter was used. This coil was driven by the standard150 V/100 A gradient power supply. In this configuration, 180 mT/m

could be reached in 180 milliseconds. As RF-coil, we used a resonator

with a 90 mm inner diameter. T2-weighed scans were acquired using a

multispin echo imaging sequence ( field of view 4 � 4 cm, 128 � 96matrix, 2.2 mm slice thickness). The tumor was determined as a region

of interest in each scan for further evaluation. The T2-relaxation time

(range 0 to >200 milliseconds) was measured in T2-map images to assess

changes in tumor tissues after treatment. It is known that necrosis andedema both have long T2 times, with necrosis even being higher than

edema. Therefore, the multispin echo sequence was used (TE = 8, 16, 24,

. . . 96 milliseconds). T2 was then calculated from these data. Tissue withT2 times between 140 and 180 milliseconds were defined as edema,

tissue with T2 times >180 milliseconds were defined as necrosis (46). The

limits were set via T2 measurements in defined regions of interest. Then,

a histogram analysis was used to quantify the relative portion of tumoredema (T2-relaxation time = 140-180 milliseconds) and necrosis (>180

milliseconds) within the entire tumor, corresponding to the measured

pixels in the regions of interest.

Immunohistochemistry. For histologic analysis, tumors were harvested

from three additional animals per treatment group, 11 days after the start of

therapy and at the end of the observation period, fixed in buffered formalin

and embedded in paraffin (42). Tissue slices (5 Am) were stained with H&E

and general tissue morphology was visualized and photographed with a

camera (Nikon Super Coolscan ED 4000, Tokyo, Japan) mounted on a Zeiss

microscope (Carl Zeiss, Jena, Germany). Tumor cell proliferation was

assessed by the percentage of Ki-67-positive cells determined by

immunohistochemical staining with the MIB-1 monoclonal mouse anti-

human Ki-67 antibody (Dako, Hamburg, Germany). Sections were counter-

stained with H&E. Ki-67 staining was quantified by counting the number of

positively stained nuclei of 200 to 250 cells in 10 randomly chosen fields

at �100 magnification. To quantify tumor vessel counts, frozen sections

were fixed and stained with primary antibody to CD31 (Becton Dickinson)

and 10 random fields at �100 magnification were chosen. To detect the

phosphorylated Akt (phospho-Akt or p-Akt) status in vivo , paraffin-

embedded tissue sections were stained using rabbit anti phospho-Akt

(Ser473) antibody (IHC specific, Cell Signaling Technology, Beverly, MA) and

Signal Stain phospho-Akt (Ser473) IHC detection Kit (Cell Signaling

Technology) according to the manufacturer’s instructions (47).

Beneficial Effects of a Trimodal Anticancer Strategy

www.aacrjournals.org 3645 Cancer Res 2005; 65: (9). May 1, 2005

Research. on February 25, 2021. © 2005 American Association for Cancercancerres.aacrjournals.org Downloaded from

Page 4: Trimodal Cancer Treatment: Beneficial Effects of Combined ... · Trimodal Cancer Treatment: Beneficial Effects of Combined Antiangiogenesis, Radiation, and Chemotherapy Peter E. Huber,

Statistical analysis. The tumor volumes V were calculated as V =0.5 � a � b2. Statistical evaluation of tumor growth was undertaken by

daily comparisons of the volumes. In addition, the general response to

treatment was assessed on the basis of the time, Tn , required to reach

n times the initial tumor volume. For multiple comparisons the Kruskall-Wallis ANOVA was used for nonparametric variables. For parametric

variables, ANOVA was used along with Fisher’s least-significant-difference.

All analyses were two-tailed. A P value of 0.05 was considered

statistically significant.

Results

Endothelial and tumor cell proliferation and clonogenicsurvival. We first investigated the effects of SU11657 alone onHUVEC and A431 cell proliferation and viability in the 72-hourproliferation/viability test. SU11657 reduced the cell number withan IC50 of 0.2 F 0.05 Amol/L for HUVEC and an IC50 of 1.5 F 0.3Amol/L for A431 (Fig. 1A). We then did combination treatments onproliferation and clonogenic survival of HUVEC and A431 tumorcells using SU11657 (0.1 Amol/L) and Pemetrexed (1.06 Amol/L)doses that had only a modest effect on proliferation inhibition ( forPemetrexed; see ref. 44). Radiation alone reduced cell proliferation

and clonogenic survival in HUVEC and tumor cells in a dose-dependent manner. The addition of either SU11657 or Pemetrexedsignificantly increased the antiproliferative effects of irradiation(P < 0.05; Fig. 1B ; Table 1) in both A431 and endothelial cells.Importantly, the triple combination resulted in a pronouncedantiproliferative effect that was more than additive and statisticallysignificant versus both dual combinations (P < 0.05) in A431 andHUVEC. This supra-additive effect is reflected by the calculatedenhancement ratios of >1 (Table 1). Interestingly, the radiosensitiz-ing drug effects were more pronounced in HUVEC than in A431cells.Similarly, for clonogenic survival as an end point, SU11657 and

Pemetrexed sensitized both A431 cells and HUVEC to radiation(P < 0.05; Fig. 1C ; Table 2). As observed for proliferation, thecombined effects were more toxic in HUVEC than in A431 cells.For example, the surviving fractions for A431 and HUVEC were0.6 versus 0.4 (at 2 Gy), 0.46 versus 0.19 (2 Gy + SU), 0.44 versus0.21 (2 Gy + Pem), 0.35 versus 0.12 (2 Gy + SU + Pem = tripletherapy). Furthermore, the more than additive anticlonogeniceffect of triple therapy was present over the entire radiationdose (0-8 Gy) in endothelial cells, whereas supra-additivity was

Figure 1. Endothelial and tumor cell proliferation and clonogenicsurvival. A, the effect of SU11657 on the proliferation of HUVECand A431 was determined by counting cells after 72 hours ofexposure to increasing concentrations of SU11657. B, the effect ofvarious treatments on the proliferation of A431 cells and HUVECwas determined by counting cells after exposure to irradiation givenalone (o) or in combination with SU11657 (E, 0.1 Amol/L),Pemetrexed (5, 1.06 Amol/L) or the triple combination (y,irradiation + Pemetrexed + SU11657). C, the clonogenic survival ofA431 and HUVEC was determined by counting colonies 14 to 21days after treatment under the same conditions as in (B). Valuesfrom combination experiments with irradiation were normalized tothe respective drug toxicity at 0 Gy. Points , mean; bars , F SD.

Cancer Research

Cancer Res 2005; 65: (9). May 1, 2005 3646 www.aacrjournals.org

Research. on February 25, 2021. © 2005 American Association for Cancercancerres.aacrjournals.org Downloaded from

Page 5: Trimodal Cancer Treatment: Beneficial Effects of Combined ... · Trimodal Cancer Treatment: Beneficial Effects of Combined Antiangiogenesis, Radiation, and Chemotherapy Peter E. Huber,

only observed for high radiation doses (>4 Gy) in A431 cells.These data suggest a high primary inherent sensitivity ofendothelial cells to trimodal treatment, but tumor cell prolifer-ation also seems to be sensitive to triple therapy.Endothelial cell migration/invasion. Endothelial cell migra-

tion and tube formation are important steps in the angiogenesisprocess. HUVEC exposure to monotherapies of 4 Gy, Pemetrexed,or SU11657 exhibited reduced migration by 20 to 30 F 4%(mean F SD) compared with the nontreated controls (P < 0.02;Fig. 2A and B). The combination of two modalities furthersignificantly inhibited the cell migration (45-65 F 6%) versuseach single therapy (P < 0.05). Triple combination resulted in a90 F 15% inhibition of HUVEC migration (P < 0.01 versus dualcombinations). These data elucidate the superior inhibitory effectof the triple combination on endothelial cell migration comparedwith each dual combination.

Endothelial tube formation. When endothelial cells such asHUVEC are plated on Matrigel, they organize into lumen-likestructures with multicentric anastomoses, as depicted 6 hoursafter plating in Fig. 2C . Exposure to monotherapies of irradiationor SU11657 alone resulted in fewer tubes and weakeranastomoses versus controls (P < 0.05), whereas Pemetrexedalone had no significant effects (P > 0.5 versus controls; Fig. 2C).Each tested dual combination led to a further reduction oftubular structures versus single therapy (P < 0.05). The triplecombination (Fig. 2C) resulted in the greatest inhibition of tube

Table 1. Radiation doses which reduce cell proliferation to60% that of controls (D60 in Gy) alone, and in combinationwith the drugs Pemetrexed and/or SU11657

Cells Treatment D60 (Gy)

F SD

Enhancement ratios

(D60Rx / D60Rx+drug)

A431 Rx 7.5 F 0.5

Rx + SU 6.6 F 0.4 1.1Rx + Pem 5.5 F 0.4* 1.4

Triple combination 4.4 F 0.3c 1.7

HUVEC Rx 4.8 F 0.3

Rx + SU 4.3 F 0.2 1.1Rx + Pem 3.6 F 0.2* 1.3

Triple combination 3.2 F 0.2c 1.5

NOTE: Enhancement ratios are defined as the quotient of D60Rx /

D60Rx + drug(s). Enhancement ratio >1 indicates greater than

independent toxicity in combined treatment.

Abbreviations: Rx, irradiation; SU, SU11657; Pem, Pemetrexed.*P < 0.05 versus Rx.cP < 0.05 versus Rx and each dual combination.

Table 2. Similar to Table 1, but for clonogenic survival asdescribed in Materials and Methods reduced to 5% byirradiation (D5 in Gy)

Cells Treatment D5 F SD

(Gy)

Enhancement ratios

(D5Rx/D5Rx + drug)

A431 Rx 6.5 F 0.4

Rx + SU 5.7 F 0.3* 1.1

Rx + Pem 5.7 F 0.3* 1.1

Triple combination 5.0 F 0.3c 1.3HUVEC Rx 5.2 F 0.3

Rx + SU 4.6 F 0.2* 1.1

Rx + Pem 4.7 F 0.2* 1.1Triple combination 4.3 F 0.2c 1.2

Abbreviations: Rx, irradiation; SU, SU11657; Pem, Pemetrexed.*P < 0.05 versus Rx.cP < 0.05 versus Rx and each dual combination.

Figure 2. Endothelial cell migration and tube formation. HUVEC migrationwas determined by counting cells that had migrated to the lower side of anuncoated invasion chamber 18 hours after cells were treated with: PBS (control),SU11657 (1.0 Amol/L), 4 Gy + Pemetrexed (1.06 Amol/L), or the triplecombination (SU11657 + 4 Gy + Pemetrexed). A, stained cells in representativefields. B, the number of HUVEC that had migrated are shown as a histogram:columns , mean; bars , F SD; Rx , 4 Gy; Pem , Pemetrexed; Su , SU11657; *,P < 0.05 versus control; **, P < 0.05 versus control and each single treatment;***, P < 0.05 versus all other treatments. C, the ability of HUVEC to formtubule-like structures when plated on Matrigel was determined for cells treatedat 6 hours after plating with: PBS (control), SU (SU11657, 1 Amol/L), 1.06 Amol/LPemetrexed (Pem), 4 Gy Radiation (Rx), and combinations. Photographs ofrepresentative fields are shown.

Beneficial Effects of a Trimodal Anticancer Strategy

www.aacrjournals.org 3647 Cancer Res 2005; 65: (9). May 1, 2005

Research. on February 25, 2021. © 2005 American Association for Cancercancerres.aacrjournals.org Downloaded from

Page 6: Trimodal Cancer Treatment: Beneficial Effects of Combined ... · Trimodal Cancer Treatment: Beneficial Effects of Combined Antiangiogenesis, Radiation, and Chemotherapy Peter E. Huber,

formation with the least number of anastomoses (P < 0.05versus dual combinations).Apoptosis in endothelial cells. Induction of apoptosis is one

possible mechanism by which the various treatments manifest

their antiproliferative effects. To explore this, the sub-G1 (apopto-tic) fractions of HUVEC (Fig. 3B) were determined by propidiumiodide fluorescence-activated cell sorting after exposure to variouscombinations of agents. Irradiation alone, Pemetrexed, or SU11657treatment alone induced 5 to 10 F 2% apoptotic cells (mean F SD,P < 0.05 versus control) at 24 hours. Exposure to each dualcombination increased the sub-G1 fractions to 10 to 20 F 3%,which was significant versus single treatments (P < 0.05). The triplecombination substantially enhanced the apoptosis rate to 25 F 4%versus all other combination (P < 0.02).To further analyze apoptosis induction, a flow cytometric

analysis of apoptotic and nonapoptotic populations of HUVECusing the anti-active caspase-3 monoclonal antibody was done.Figure 3C shows representative results at 6 hours after treatment,demonstrating that untreated control endothelial cells wereprimarily negative for caspase-3; a slight shift of the curves wasobserved after radiation and Pemetrexed single therapy, indicatingsome caspase-3 activity. SU11657 (1 Amol/L) induced morecaspase-3 activity than radiation (4 Gy) or Pemetrexed (1.06Amol/L) at the concentrations chosen. Dual combinations resultedin more caspase-3 activity than monotherapies, and triplecombination markedly shifted the curve further to the rightindicating the most caspase-3 activity. Thus, triple combinationmarkedly induced endothelial cell apoptosis.Akt phosphorylation in endothelial cells in vitro . Akt is

thought to contribute to survival signaling mediated by receptortyrosine kinases such as VEGFR2 and PDGFR. Therefore, theextent of Akt activation was examined after exposure to varioustreatments (Fig. 3C and D). Although irradiation or Pemetrexedincreased apoptosis, as single agents or in combination, thesetreatments were also observed to increase Akt phosphorylationsuggesting the induction of a cell survival mechanism in responseto toxicity. In contrast, SU11657 attenuated Akt activity andsuppressed the enhancement of Akt phosphorylation observedafter irradiation or exposure to the chemotherapeutic agent. Wealso found that the combination of radiation and chemotherapyresulted in the highest Akt phosphorylation. Interestingly,

Figure 3. Endothelial cell apoptosis and Akt signaling. A, the percentageof apoptotic HUVEC was determined by flow cytometric analysis of cellswith sub-G1 DNA content at 24 hours after different treatments: Rx, 4 Gy;Pem, 1.06 Amol/L; SU11657, 1.0 Amol/L. Columns , mean; bars , F SD.Statistical significance is indicated by: *, P < 0.05 versus control; **,P < 0.05 versus control and single treatments; ***, P < 0.05 versus allother treatments. B, flow cytometric analysis of apoptotic andnonapoptotic endothelial cells (HUVEC) using a PE-conjugated activecaspase-3 monoclonal antibody at 6 hours after therapy. Control cellsare primarily negative for the presence of active caspase-3. SU11657(1 Amol/L) induces more caspase-3 activity than irradiation (4 Gy) orPemetrexed (1.06 Amol/L). Dual combinations induce more caspase-3activity than monotherapies. After triple combination, the most number ofcells are positive for active caspase-3 staining. C, activated Akt was assessedby immunocytochemistry of phosphorylated Akt protein. HUVEC weretreated with SU11657 for 2 hours (SU, 1 Amol/L), or treated withPemetrexed (Pem, 1.06 Amol/L) for 2 hours, or irradiated (Rx, 4 Gy), ortreated with a combination of Pemetrexed and SU11657 for 2 hoursand irradiated (Triple). Immediately after treatment, cells were incubatedon coverslips for 45 minutes and stained with a specific antibodyagainst phosphorylated Akt Ser473 (green ). For nuclear localization, cellswere counterstained with propidium iodide (red ). Representative fieldsare shown. D, quantitative image analysis of phosphorylated Akt afterstaining with Akt-specific antibody in HUVEC 45 minutes after the end oftherapy as described in Methods and Materials. Columns , mean normalizedto control; bars , F SD of averaged intensity of phospho-Akt signals of10 microscope fields, each depicting 15 to 30 endothelial cells; *, significant(P < 0.05).

Cancer Research

Cancer Res 2005; 65: (9). May 1, 2005 3648 www.aacrjournals.org

Research. on February 25, 2021. © 2005 American Association for Cancercancerres.aacrjournals.org Downloaded from

Page 7: Trimodal Cancer Treatment: Beneficial Effects of Combined ... · Trimodal Cancer Treatment: Beneficial Effects of Combined Antiangiogenesis, Radiation, and Chemotherapy Peter E. Huber,

SU11657 down-regulated radiation-, chemotherapy-, and radio/chemotherapy-induced Akt phosphorylation significantly in hu-man endothelial cells (P < 0.05; Fig. 3D).Tumor growth of A431 xenografts in nude mice. We then

investigated the combined effects of irradiation, chemotherapywith Pemetrexed, and antiangiogenic therapy with SU11657 in vivoin A431 tumor xenografts growing s.c. on hind limbs of BALB/cnude mice. Because radiotherapy (7.5 Gy) was to be given onlyonce, and the optimal schedule of combining antiangiogenictherapy and radiotherapy was in particular unknown, we firstasked whether the order of administration of radiotherapy andangiogenesis inhibition influenced tumor growth. We found thatmonotherapies with either radiotherapy or SU11657 significantlyattenuated tumor growth versus vehicle control (P < 0.05, fromday 7 onwards; Fig. 4A). Furthermore, both schedules of combinedantiangiogenic and radiotherapy were more efficacious than

monotherapies from day 12 onwards (P < 0.05). Importantly, wefound that when radiotherapy was given 1 day after startingSU11657 therapy (SU + Rx), tumor growth delay was greater thanwhen radiotherapy preceded SU11657 (Rx + SU; P < 0.05, from day12 onwards).Therefore, for the following trimodal tumor growth experi-

ments, we chose the more efficacious schedule with SU11657administration starting prior to radiotherapy. Here, mice withestablished tumors were stratified into nine therapy arms (Fig.4B). We observed reduced tumor growth in each treatment armversus control (Fig. 4C) from day 7 onwards (P < 0.05 versuscontrols from day 7 onward, by day comparison). Tumor growthdelay was significantly increased in each dual combinationcompared with monotherapies (P < 0.05, from day 9 onward).The triple combination significantly further reduced tumorgrowth compared with dual therapies ( from day 11 onwards,

Figure 4. In vivo growth of A431 tumor xenografts. Mice with A431 human epidermoid cell carcinoma growing s.c. on BALB/c nu /nu mice were treated withirradiation (Rx, 7.5 Gy), with the chemotherapeutic agent Pemetrexed (150 mg/kg for 4 consecutive days starting at day 0, 600 mg total dose) and/or withSU11657 (100 mg/ kg thrice weekly until end of observation). A second triple therapy regimen (Triple metron) with a metronomic Pemetrexed administration oversix injections until day 10 was introduced (100 mg/kg, 600 mg total dose). Abbreviations: Rx, 7.5 Gy; Pem, Pemetrexed (150 mg/kg); SU, SU11657 (100 mg/kg);Triple, SU + Pem + Rx (150 mg/kg � 4); Triple metron, SU + Pem + Rx (100 mg/kg � 6). The tumor growth of s.c. A431 tumors in BALB/c nu /nu mice is shown,with each treatment group containing 10 to 12 mice, and the data points representing the mean F SE of tumor volumes. A, tumor volumes after radiotherapy andangiogenesis inhibition to investigate the role of the schedule of administration. Irradiation 1 day after start of SU11657 therapy (SU + Rx) was more effective thanirradiation 1 day prior to SU11657 start (Rx + SU; #, P < 0.05 from day 12 onwards). Monotherapy significantly attenuates tumor growth from day 7 onwards (*P < 0.05).Both schedules of dual irradiation and SU11657 also result in more tumor growth delay than each respective single therapy from day 12 onwards (**P < 0.05 foreach comparison). B, schedule diagram for the nine groups in the trimodal therapy experiment. The more effective strategy from (A) is employed with SU11657starting prior to irradiation. Two chemotherapy arms are tested, one conventional arm and one metronomic arm. C, tumor growth for the eight groups in thetrimodal therapy experiment. The by day comparison shows significant differences for all treatment groups versus control from day 7 onwards (*P < 0.05). Eachcombination of two therapies resulted in more tumor growth delay than each respective monotherapy from day 9 onwards (**P < 0.05 for each comparison). Both tripleschedules resulted in more tumor growth delay than all other groups, which was significant from day 11 on (***P < 0.05). The triple metronomic chemotherapywas significantly more effective than conventional chemotherapy therapy (#P < 0.05, from day 21 onwards). D, the tumor growth delay was also derived from T5,which represents the time required to reach five times the initial tumor volume, as an overall measure of tumor growth dynamics. Columns , difference in T5 of therespective treatment group and T5 in control group F SD. Statistical significance is designated as: *, significant (P < 0.05) versus control; **, significant (P < 0.05) versuscontrol and each single treatment group; ***, significant (P < 0.05) versus control, each single and double treatment group.

Beneficial Effects of a Trimodal Anticancer Strategy

www.aacrjournals.org 3649 Cancer Res 2005; 65: (9). May 1, 2005

Research. on February 25, 2021. © 2005 American Association for Cancercancerres.aacrjournals.org Downloaded from

Page 8: Trimodal Cancer Treatment: Beneficial Effects of Combined ... · Trimodal Cancer Treatment: Beneficial Effects of Combined Antiangiogenesis, Radiation, and Chemotherapy Peter E. Huber,

Figure 5. Immunohistochemistry of A431 tumors. Tumor cell proliferation and microvessel density were assessed by Ki-67 and CD31 immunostaining in A431tumors excised on day 11 after start of therapy with: no treatment (control tumor), radiation (Rx, 7.5 Gy), Pem (Pemetrexed), SU (SU11657), SU + Rx and the reverseschedule Rx + SU as shown in Fig. 4, Rx + Pem, SU + Pem + Rx (conventional triple therapy and metronomic chemotherapy dosing as shown in Fig. 4). A,representative fields of Ki-67 staining are shown. B, quantitative analysis of Ki-67-positive cells determined by counting immunostained cells in tumors treated with:*, significant (P < 0.05) versus control; **, significant (P < 0.05) versus control and each monotherapy; ***, significant (P < 0.05) versus all other groups. C,representative fields of CD31 immunostaining in A431 tumor. D, quantitative analysis of CD31-positive vessels in the center of A431 tumors: *, significant (P < 0.05)versus control; **, significant (P < 0.05) versus control and respective monotherapies; ***, significant (P < 0.05) versus all other groups, #, significant (P < 0.05)versus conventional triple therapy. x, significant between SU + Rx versus Rx + SU (P < 0.05). E, representative figures of phosphorylated Akt (p-Akt) immunostainingin A431 tumors after indicated treatments. Paraffin-embedded tissue sections of control (Ctrl), SU11657 (SU), Pemetrexed (Pem), radiation (Rx), radiation +Pemetrexed (Rx + Pem) and triple combination (radiation + Pemetrexed + SU11657, Triple) were stained using rabbit anti phospho-Akt (Ser473) antibody. Left column ,�100 view; right column , insets of the �100 view at �400. The black arrows point to endothelial cells. *, erythrocytes within the vessels.

Cancer Research

Cancer Res 2005; 65: (9). May 1, 2005 3650 www.aacrjournals.org

Research. on February 25, 2021. © 2005 American Association for Cancercancerres.aacrjournals.org Downloaded from

Page 9: Trimodal Cancer Treatment: Beneficial Effects of Combined ... · Trimodal Cancer Treatment: Beneficial Effects of Combined Antiangiogenesis, Radiation, and Chemotherapy Peter E. Huber,

P < 0.05). Interestingly, the metronomic triple chemotherapydosing was slightly but significantly more effective than theconventional triple chemotherapy schedule (P < 0.05, from day 21onwards).

In addition to the by day comparison, the tumor growth delaywas assessed on the basis of the time, T5, required to reach fivetimes the initial tumor volume (Fig. 4D). In principle, agreementwith the above by day comparison, single treatments induced a

Figure 6. In vivo functional analysisby MRI and IFP measurements.T2-weighed MRI of A431 tumors growings.c. on hind limbs of BALB/c nu /numice was done to assess tumor size,necrosis, and edema on day 10 afterstart of therapy. Tumor borders areoutlined to indicate the region ofinterest. Closed arrows , regions ofnecrosis; open arrows , regions ofedema. A, representative T2-weighedtransversal scans of the tumors. B,representative parameter images of thedistribution of the T2 relaxation time.The T2-map scans enable calculationof the T2-relaxation times of tumortissue, tumor necrosis, and edema. C,quantitative analysis of T2-map scansis shown: T2-relaxation times of 140 to180 milliseconds correspond to regionsof edema and those >180 millisecondsrepresent regions of necrosis. D, IFPwas measured in A431 tumorxenografts on day 1, at 24 hours afterstart of the respective therapy, asshown in Fig. 5. IFP was significantlydecreased after treatment withSU11657. Columns , mean; bars ,F SD; *, significant (P <0.05) versuscontrol, irradiation alone andPemetrexed alone.

Beneficial Effects of a Trimodal Anticancer Strategy

www.aacrjournals.org 3651 Cancer Res 2005; 65: (9). May 1, 2005

Research. on February 25, 2021. © 2005 American Association for Cancercancerres.aacrjournals.org Downloaded from

Page 10: Trimodal Cancer Treatment: Beneficial Effects of Combined ... · Trimodal Cancer Treatment: Beneficial Effects of Combined Antiangiogenesis, Radiation, and Chemotherapy Peter E. Huber,

significant tumor growth delay versus control of 4.1 F 1 days(radiation), 3.5 F 1 days (Pemetrexed), or 1.8 F 0.5 days (SU11657),respectively (P < 0.05 for all three). Each of the dual combinationsproduced a further significant tumor growth delay compared withsingle treatments (P < 0.05, each). Triple combination markedlyenhanced tumor growth delay versus all other groups (9.7 F 1.5days, P < 0.02 versus dual combinations).All therapies were well-tolerated by the animals. No differences in

animal weight or clinical behavior between groups were detected.All animals survived until the end of the observation period unlesssacrificed for scheduled histology. Furthermore, histology in HE-stained sections from organs including liver, spleen, kidney, skin,and muscle did not reveal therapy-related toxicity.Histopathology of A431 tumors. In order to assess tumor cell

proliferation under different treatment regimens, the Ki-67 indexwas determined (Fig. 5A). The total number of Ki-67-positive A431cells was reduced in all treated groups versus control. Dualcombinations had lower Ki-67 indices than single therapies.Significantly, the triple combination resulted in a marked furtherdecrease of the Ki-67 index versus all dual combinations (P < 0.05;Fig. 5B). In addition, the tumor cells exhibited phenotypic andmorphologic alterations, such as swollen cytoplasm, which wasmost pronounced after triple-therapy. When comparing theschedule of radiation and antiangiogenic treatment from thetumor growth experiments shown in Fig. 4A , antiangiogenic priorto radiation (SU + Rx versus Rx + SU) had less Ki-67-positive cells,but without statistical significance.Vessel density in the tumors was determined by CD31 staining

(Fig. 5C). A significant decrease in vessel count was observed aftersingle treatment with SU11657 or irradiation (P < 0.05), whereastreatment with Pemetrexed alone had no significant influence onvessel count (P > 0.8). Combining SU11657 with irradiation furtherreduced vessel count compared with single treatments (P < 0.05).Combining Pemetrexed with irradiation reduced vessel density to acomparable amount as irradiation alone, as expected based on thelack of activity of Pemetrexed alone. However, when Pemetrexedwas combined with SU11657, fewer vessels were observed thanwith SU11657 alone. The triple combination caused the greatestantiangiogenic effects in vivo , with the lowest vessel number versusall other groups (P < 0.05; Fig. 5D). When comparing the schedule

of radiation and antiangiogenic treatment, antiangiogenic treat-ment prior to radiation caused significantly less vessel density than(P < 0.05, SU + Rx versus Rx + SU).In vitro in endothelial cells, we had shown (Fig. 3C and D) that

Akt was phosphorylated after radio/chemotherapy and SU11657attenuated this process. To transfer the in vitro results immuno-histochemistry was done in treated A431 tumors. We found thatradiotherapy, chemotherapy, and their combination induced Aktphosphorylation which was attenuated by SU11657, particularlyvisible after combined radio/chemotherapy. This is highlighted inFig. 5E , showing phosphorylated Akt (p-Akt) immunostaining incontrol (Ctrl), SU11657 (SU), Pemetrexed (Pem), radiation (Rx),radiation + Pemetrexed (Rx + Pem) and triple combination(radiation + Pemetrexed + SU11657, Triple) treated A431 tumors(Fig. 5E).Morphologic and functional magnetic resonance imaging of

A431 tumors. To obtain additional functional insights into thetumor physiology, MRI T2-weighed images were captured on day 10after therapy began. The first column in Fig. 6A illustrates thetumor growth delay with dual therapies being more effective thansingle therapies, and triple therapies showing the greatest tumorgrowth inhibition. In addition, a reduction of tumor infiltrationinto the adjacent muscle tissue is shown, in particular, after dualand triple combination. MRI also enabled a quantitative assess-ment of tumor necrosis and edema using the pixelwise analysis, asdepicted in T2-maps (Fig. 6B). The histogram analysis (Fig. 6C)represents quantification of the relative amount of tumor edema(T2-relaxation time, 140-180 milliseconds) and necrosis (T2 >180milliseconds) within the area depicted as tumor. Of note, tumorstreated with SU11657 in dual combinations contained a lowerpercentage of pixels assigned as edema than respective mono-therapies (Table 3). This edema-reductive effect of SU11657 wasparticularly visible in combination with Pemetrexed, radiotherapyand in triple combination as shown in Table 3. The table alsoshows that SU11657 + Pem + Rx triple therapy had the highestpercentage of tumor pixels, and at the same time, the leastpercentage of edema and necrosis.Interstitial fluid pressure in A431 tumors. Because IFP has

been suggested to be a critical factor in tumor perfusion, edemaformation, and in other variables relevant to the tumor

Table 3. Quantitative histogram analysis of MRI T2-maps as the number of pixels indicating tumor, edema, and necrosis intwo animals at day 10

Treatment group % (mean F SE) of all pixels in tumor volume

Tumor tissue Tumor edema Tumor necrosis

Control 93 F 2.5 4 F 1 3 F 0SU 91 F 0 4 F 1 5 F 2

Pem 77 F 4 15 F 3 8 F 3

Rx 87 F 4.5 9 F 0 4 F 3

SU + Rx 94 F 2* 4 F 1* 2 F 1.5*Pem + Rx 93 F 1.5 4 F 0 3 F 1

Pem + SU 97 F 1* 2 F 1* 1 F 1*

SU + Pem + Rx 99 F 0c 1 F 0c 0 F 0c

Abbreviations: Rx, irradiation; SU, SU11657; Pem, Pemetrexed.*P < 0.05 for dual combinations containing SU versus respective monotherapies.cP < 0.05 for triple versus all other except for triple versus Pem + SU.

Cancer Research

Cancer Res 2005; 65: (9). May 1, 2005 3652 www.aacrjournals.org

Research. on February 25, 2021. © 2005 American Association for Cancercancerres.aacrjournals.org Downloaded from

Page 11: Trimodal Cancer Treatment: Beneficial Effects of Combined ... · Trimodal Cancer Treatment: Beneficial Effects of Combined Antiangiogenesis, Radiation, and Chemotherapy Peter E. Huber,

pathophysiology, we determined the IFP in A431 tumors in vivo . IFPwas measured 1 day after the beginning of each monotherapy todetermine the condition for the subsequent therapies which werescheduled for that day. SU11657 markedly decreased the IFP intumors 1 day after institution. The IFP decreased significantly from8.8 F 2.6 for controls to 4.2 F �1.5 mm Hg (mean F SD, P < 0.02)with SU11657. In contrast, radiotherapy and chemotherapy alonehad no significant effects on IFP (P > 0.5; Fig. 6D). Tumor size alsoinfluenced IFP. Figure 6D illustrates that for control tumors withincreasing tumor size, IFP increased from 8.8F 2.6 mm Hg on day 1to 12.7F 2.8 on day 3 (P < 0.01). SU11657-treated tumors, alone or incombination with other therapies, had significantly lower IFPvalues than controls (P < 0.05), and triple therapy resulted in smallerIFP than monotherapies (P < 0.05). Figure 6D also shows the IFPresults from tumors of the radiotherapy/SU11657 schedulingexperiments (Fig. 4A). The regimen with radiotherapy given afterinstitution of SU11657 (SU + Rx) had lower IFP than the schedulewith radiotherapy prior to SU11657 (Rx + SU), although thisdifference did not reach statistical significance (P > 0.1).

Discussion

Here we report on the beneficial combination of a trimodalanticancer strategy consisting of radiotherapy, chemotherapy, andangiogenesis inhibition in vitro and in vivo . We present molecular,cellular, and physiologic evidence for the beneficial use of thismultimodal combination. Due to the clinical availability of thethree modalities, including angiogenesis inhibitors such as VEGFantibodies, it is possible that this or a similar trimodal therapyregimen can be investigated in a variety of clinical entities.One reason for the beneficial trimodal combination could be

that SU11657 interferes with the radio/chemotherapy-associatedparacrine tumor cell-endothelial cell interaction. For example, ithas been shown that ionizing radiation exerts both proangiogenicand antiangiogenic effects (9). Whereas irradiation of endothelialcells has antiproliferative and proapoptotic effects, irradiation ofthe tumor cell compartment can increase the expression of keyproangiogenic cytokines such as VEGF and basic fibroblast growthfactor (9, 11). Irradiation of endothelial cells may also induceexpression of PDGF (48). Thus, radiation may induce paracrineproangiogenic effects which have been reported to ultimatelyreduce the radiosensitivity of tumor endothelium (9, 11). BecauseSU11657 inhibits VEGF and PDGF signaling, SU11657 may be ableto attenuate this escape mechanism and thereby enhance theantiendothelial radiation effects.Another reason for the efficacious trimodal combination might

be their opposite effects on Akt signaling. Akt might beimportant here, because Akt is a cytoplasmic serine/threoninekinase that, among other things, mediates survival signalingfrom receptor tyrosine kinases such as VEGFR and PDGFR (36).It is activated in many cancers and may promote drug resistance(34, 35). Our data show that irradiation and/or exposure toPemetrexed induces Akt phosphorylation, a surrogate foractivation in endothelial cells in vitro and in vivo , and we alsoshow that SU11657 attenuates this Akt activation. It is likely thatthis occurs through interruption of autocrine signaling. Wefound in functional angiogenesis assays that SU11657 enhancesthe sensitivity of endothelial cells to radiation or radio/chemotherapy, presumably by attenuating Akt activation. It hasbeen suggested that microvascular damage mediates thesensitivity of tissues to radiotherapy in general (49). On a

molecular and cellular level, it is thus plausible that endothelialcells are directly resensitized by SU11657 to radio/chemotherapy.Cell cycle and caspase-3 activity assessment further showed thatapoptosis played an important role in the reaction of endothelialcells to triple combination.Aside from the effects on the endothelial cell compartment, we

also found that triple therapy was remarkably effective directlyagainst A431 cells. In vitro proliferation or clonogenic survivalassays showed that A431 tumor cells were sensitive to SU11657alone and in combination with irradiation or Pemetrexed. Thismight suggest that SU11657 interferes with autocrine proliferationmediated by split kinases, but it is more likely to inhibit anotherkinase that contributes to A431 proliferation and survival. Theidentity of this kinase is not currently known. EGFR, which isknown to be overexpressed and activated in A431 cells, is notinhibited by SU11657 (IC50 > 20 Amol/L), rendering it an unlikelycandidate.An unresolved issue in multimodal therapy, both experimentally

and clinically, is the issue of the optimal treatment scheduling. Formany cancer types, concurrent or sequential, and continuous orbolus type applications are pursued. Although combining anti-angiogenic therapy and radiotherapy has been shown empiricallyto be beneficial in experimental tumor systems, their optimalscheduling is in particular unclear, due to issues of a potentialshutdown of blood vessels, inducing hypoxia and thus reducingradiosensitivity. In our A431 tumor system in mice, we found thatwhen single-dose radiotherapy was given after starting the SU11657therapy, tumor growth delay was greater, and microvessel densitywas reduced versus the schedule when radiotherapy precededSU11657 therapy. Our data thus suggest that radiotherapy is moreeffective when tumors are pretreated with antiangiogenic therapy.However, both schedules of combined dual antiangiogenic andradiotherapy reduced tumor growth more efficacious than eachrespective monotherapy. Most important to us, trimodal therapywith additional chemotherapy clearly resulted in the greatesttumor growth delay in the A431 xenograft model. Remarkably, ofthe two trimodal arms tested, metronomic chemotherapy (lowerdaily, but the same total dosage) was more efficacious in tumorgrowth delay than the conventional arm, in particular towards theend of observation.This therapeutic enhancement by the trimodal therapy was

accompanied by a significantly decreased Ki-67 proliferationindex and reduced vascular density versus all other groups. Ofnote, at the histology assay time, tumors in some groups haddifferent sizes, and thus their differences in histology may not beexclusively treatment-related, but are additionally size-dependent.However, when tumors were excised at day 11, no significantdifferences in tumor size between conventional and metronomicchemotherapy dosing were apparent yet, whereas histology wasdifferent, suggesting an inherent treatment-related effect: metro-nomic administration of Pemetrexed caused reduced tumormicrovessel density compared with the conventional Pemetrexedregimen. Later in the course of the observation, this histologicdifference in CD31 count translated into a tumor size difference.Together with published antiangiogenic effects of metronomicchemotherapy dosing (1, 50), we conclude from our data thatthe antitumor effects after trimodal therapy partially result fromthe combined enhancement of antiangiogenic effects.Another important issue for multimodal cancer therapy is

treatment-related toxicity. Despite the better antitumor effects inthe trimodal groups, we did not observe an increase of the overall

Beneficial Effects of a Trimodal Anticancer Strategy

www.aacrjournals.org 3653 Cancer Res 2005; 65: (9). May 1, 2005

Research. on February 25, 2021. © 2005 American Association for Cancercancerres.aacrjournals.org Downloaded from

Page 12: Trimodal Cancer Treatment: Beneficial Effects of Combined ... · Trimodal Cancer Treatment: Beneficial Effects of Combined Antiangiogenesis, Radiation, and Chemotherapy Peter E. Huber,

toxicity as determined by clinical parameters or in histology in avariety of organs. However, it is obvious that adverse effects cannotbe completely excluded, particularly in case of the translation inthe clinical setting.Apart from direct cellular effects, combination treatments also

influence general tumor physiology such as IFP. Elevated IFP is ahallmark of solid tumors that emerges due to the expansive growthcharacteristic of the tumors and their abnormal vasculararchitecture (3, 12, 19, 39). We found that the IFP in A431 tumorswas significantly reduced by f50% at 24 hours after SU11657administration. One explanation would be that SU11657 decreasesvascular permeability via inhibition of VEGFR or PDGFR, which inturn may lower IFP. In contrast, both radiation and chemotherapydid not substantially alter the IFP. Likewise, quantitative T2-weighed MRI showed that the IFP reduction was accompanied byreduced intratumoral edema. MRI parameters may thus be used assurrogate markers to predict the efficacy of antiangiogenic therapyearly in the treatment course.From these physiology data, we found that in our trimodal therapy

study, radiotherapy was delivered at reduced IFP 1 day after the startof SU11657 treatment. Thus, the reduction of tumor IFP and edemaoffers one physiologic indication for the improved efficacy ofradiotherapy or chemotherapy when given after SU11657. This viewis supported by our data showing that changing the order of therapyand giving SU11657 after radiotherapy was less effective in reducingtumor growth than vice versa.This concept of increased radio/chemotherapy efficacy

through normalization of tumor vasculature by SU11657inhibition is consistent with preclinical and clinical observationsfrom other strategies targeting the VEGF pathway (3, 11, 12, 19,24, 38–41). In agreement with our data using SU11657, aprevious study has shown that SU11248 (a chemical analogue ofSU11657) inhibited VEGFR signaling and led to reduced vascularpermeability in mice (22). On the other hand, it has been shownthat treatment with a PDGF receptor tyrosine kinase inhibitor,STI571 (Gleevec), also decreased the interstitial hypertension andincreased capillary-to-interstitium transport of 5-fluorouracil inan experimental colon carcinoma model (51, 52). Therefore, it isconceivable that the combined VEGF and PDGF signalinginterruption by the receptor tyrosine kinase inhibitor SU11657is a powerful tool to normalize the tumor vasculature (53, 54).Likewise, the inhibition of both VEGF and PDGF signaling usingSU11248, an analogue to SU11657, has recently been reported to

produce survival benefits in combination with metronomicchemotherapy schedule as a beneficial anticancer strategy inpancreatic islet tumors of mice (55).On the underlying signaling level, we found that SU11657

inhibited Akt-mediated survival signals in vitro in HUVEC andin vivo in the A431 tumor model. Akt is considered a majorescape mechanism from tumor therapy (33–37, 44). In the contextof the proposed trimodal therapy, our results suggest a dual rolefor Akt, which is downstream from VEGF/PDGF, and thus alsorepresents a link to decreased IFP after VEGF/PDFG inhibition(56). First, the inhibition of Akt signaling can contribute to thebeneficial physiologic effects for the subsequent radio/chemo-therapy (and may indirectly affect other ‘‘normal’’ and tumorcells). Second, Akt inhibition by SU11657 also represents amechanism by which the individual endothelial cell is directlyresensitized to radio/chemotherapy, which is otherwise disturbedby the up-regulation of Akt as a self defense to cell toxicity.It should be noted that similar experiments have not yet been

done with spontaneously arising tumors. Because the vasculatureof s.c. tumor xenografts may not accurately reflect the vasculatureof tumors growing in other organs or tissues, it has yet to be shownthat all of the effects of triple therapy reported here will also beobserved in more natural tumors.Current approaches in clinical cancer therapy favor multimodal

strategies (3–6). One rationale is the concept that the side effects ofdifferent therapies do not overlap. The combination of irradiationand chemotherapy has become a standard treatment and isassociated with improved survival rates in many tumors (2). Thesimultaneous combination of chemotherapy and antiangiogenesishas shown clinical promise (4–6). We suggest that, given thedisparate modes of action, the proposed triple combination ofirradiation, chemotherapy, and antiangiogenesis using a VEGF/PDGF receptor antagonist has clinical potential as an anticancerstrategy.

Acknowledgments

Received 5/19/2004; revised 2/16/2005; accepted 2/25/2005.The costs of publication of this article were defrayed in part by the payment of page

charges. This article must therefore be hereby marked advertisement in accordancewith 18 U.S.C. Section 1734 solely to indicate this fact.

We thank Thuy Trinh, Ute Haner, Cornelia Leimert, and Alexandra Tietz forexcellent technical assistance; Drs. G. McMahon and A. Howlett for support; and Profs.Klaus-Josef Weber for support with FACS measurements and Michael Wannenmacherfor general support.

References1. Browder T, Butterfield CE, Kraling BM, et al.Antiangiogenic scheduling of chemotherapy improvesefficacy against experimental drug-resistant cancer.Cancer Res 2000;60:1878–86.

2. Choy H, Kim DW. Chemotherapy and irradiationinteraction. Semin Oncol 2003;30:3–10.

3. Jain RK. Normalizing tumor vasculature with anti-angiogenic therapy: a new paradigm for combinationtherapy. Nat Med 2001;7:987–9.

4. Kerbel R, Folkman J. Clinical translation ofangiogenesis inhibitors. Nat Rev Cancer 2002;2:727–39.

5. Fernando NH, Hurwitz HI. Inhibition of vascularendothelial growth factor in the treatment of colorectalcancer. Semin Oncol 2003;30:39–50.

6. Kabbinavar F, Hurwitz HI, Fehrenbacher L, et al. PhaseII, randomized trial comparing bevacizumab plus

fluorouracil (FU)/leucovorin (LV) with FU/LV alone inpatients with metastatic colorectal cancer. J Clin Oncol2003;21:60–5.

7. Yang JC, Haworth L, Sherry RM, et al. A randomizedtrial of bevacizumab, an anti-vascular endothelialgrowth factor antibody, for metastatic renal cancer.N Engl J Med 2003;349:427–34.

8. Burke PA, DeNardo SJ. Antiangiogenic agents andtheir promising potential in combined therapy. Crit RevOncol Hematol 2001;39:155–71.

9. Abdollahi A, Lipson KE, Han X, et al. SU5416 andSU6668 attenuate the angiogenic effects of radiation-induced tumor cell growth factor production andamplify the direct anti-endothelial action of radiationin vitro . Cancer Res 2003;63:3755–63.

10. Geng L, Donnelly E, McMahon G, et al. Inhibition ofvascular endothelial growth factor receptor signalingleads to reversal of tumor resistance to radiotherapy.Cancer Res 2001;61:2413–9.

11. Gorski DH, Beckett MA, Jaskowiak NT, et al. Blockageof the vascular endothelial growth factor stressresponse increases the antitumor effects of ionizingradiation. Cancer Res 1999;59:3374–8.

12. Lee CG, Heijn M, di Tomaso E, et al. Anti-vascularendothelial growth factor treatment augments tumorradiation response under normoxic or hypoxicconditions. Cancer Res 2000;60:5565–70.

13. Chan LW, Camphausen K. Angiogenic tumormarkers, antiangiogenic agents and radiation therapy.Expert Rev Anticancer Ther 2003;3:357–66.

14. Ma BB, Bristow RG, Kim J, Siu LL. Combined-modality treatment of solid tumors using radiotherapyand molecular targeted agents. J Clin Oncol 2003;21:2760–76.

15. O’Reilly MS. Targeting multiple biological pathwaysas a strategy to improve the treatment of cancer. ClinCancer Res 2002;8:3309–10.

Cancer Research

Cancer Res 2005; 65: (9). May 1, 2005 3654 www.aacrjournals.org

Research. on February 25, 2021. © 2005 American Association for Cancercancerres.aacrjournals.org Downloaded from

Page 13: Trimodal Cancer Treatment: Beneficial Effects of Combined ... · Trimodal Cancer Treatment: Beneficial Effects of Combined Antiangiogenesis, Radiation, and Chemotherapy Peter E. Huber,

16. Siemann DW, Shi W. Targeting the tumor bloodvessel network to enhance the efficacy of radiationtherapy. Semin Radiat Oncol 2003;13:53–61.

17. Wachsberger P, Burd R, Dicker AP. Tumor responseto ionizing radiation combined with antiangiogenesisor vascular targeting agents: exploring mechanisms ofinteraction. Clin Cancer Res 2003;9:1957–71.

18. Colevas AD, Brown JM, Hahn S, Mitchell J,Camphausen K, Coleman CN. Development of inves-tigational radiation modifiers. J Natl Cancer Inst 2003;95:646–51.

19. Willett CG, Boucher Y, di Tomaso E, et al. Directevidence that the VEGF-specific antibody bevacizumabhas antivascular effects in human rectal cancer. NatMed 2004;10:145–7.

20. Cain JA, Grisolano JL, Laird AD, Tomasson MH.Complete remission of TEL-PDGFRB-induced myelo-proliferative disease in mice by receptor tyrosine kinaseinhibitor SU11657. Blood 2004;104:561–4.

21. De Bandt M, Ben Mahdi MH, Ollivier V, et al.Blockade of vascular endothelial growth factor receptorI (VEGF-RI), but not VEGF-RII, suppresses jointdestruction in the K/BxN model of rheumatoidarthritis. J Immunol 2003;171:4853–9.

22. Mendel DB, Laird AD, Xin X, et al. In vivo antitumoractivity of SU11248, a novel tyrosine kinase inhibitortargeting vascular endothelial growth factor andplatelet-derived growth factor receptors: determinationof a pharmacokinetic/pharmacodynamic relationship.Clin Cancer Res 2003;9:327–37.

23. O’Farrell AM, Foran JM, Fiedler W, et al. An innovativephase I clinical study demonstrates inhibition of FLT3phosphorylation by SU11248 in acute myeloid leukemiapatients. Clin Cancer Res 2003;9:5465–76.

24. Schueneman AJ, Himmelfarb E, Geng L, et al.SU11248 maintenance therapy prevents tumor regrowthafter fractionated irradiation of murine tumor models.Cancer Res 2003;63:4009–16.

25. Sohal J, Phan VT, Chan PV, et al. A model of APL withFLT3 mutation is responsive to retinoic acid and areceptor tyrosine kinase inhibitor, SU11657. Blood 2003;101:3188–97.

26. Verheul HM, Pinedo HM. Vascular endothelialgrowth factor and its inhibitors. Drugs Today (Barc)2003;39 Suppl C:81–93, 81–93.

27. Chen VJ, Bewley JR, Andis SL, et al. Cellularpharmacology of MTA: a correlation of MTA-inducedcellular toxicity and in vitro enzyme inhibition with itseffect on intracellular folate and nucleoside triphos-phate pools in CCRF-CEM cells. Semin Oncol 1999;26:48–54.

28. Hanauske AR, Chen V, Paoletti P, Niyikiza C.Pemetrexed disodium: a novel antifolate clinicallyactive against multiple solid tumors. Oncologist 2001;6:363–73.

29. Sischy B. The use of radiation therapy combined withchemotherapy in the management of squamous cellcarcinoma of the anus and marginally resectableadenocarcinoma of the rectum. Int J Radiat Oncol BiolPhys 1985;11:1587–93.

30. Bischof M, Weber KJ, Blatter J, Wannenmacher M,Latz D. Interaction of pemetrexed disodium (ALIMTA,multitargeted antifolate) and irradiation in vitro . Int JRadiat Oncol Biol Phys 2002;52:1381–8.

31. Bischof M, Huber P, Stoffregen C, Wannenmacher M,Weber KJ. Radiosensitization by pemetrexed of humancolon carcinoma cells in different cell cycle phases. Int JRadiat Oncol Biol Phys 2003;57:289–92.

32. Teicher BA, Chen V, Shih C, et al. Treatmentregimens including the multitargeted antifolateLY231514 in human tumor xenografts. Clin Cancer Res2000;6:1016–23.

33. Hanahan D, Weinberg RA. The hallmarks of cancer.Cell 2000;100:57–70.

34. Wendel HG, De Stanchina E, Fridman JS, et al.Survival signalling by Akt and eIF4E in oncogenesis andcancer therapy. Nature 2004;428:332–7.

35. Johnstone RW, Ruefli AA, Lowe SW. Apoptosis: a linkbetween cancer genetics and chemotherapy. Cell 2002;108:153–64.

36. Edwards E, Geng L, Tan J, Onishko H, Donnelly E,Hallahan DE. Phosphatidylinositol 3-kinase/Akt sig-naling in the response of vascular endothelium toionizing radiation. Cancer Res 2002;62:4671–7.

37. McKenna WG, Muschel RJ. Targeting tumor cells byenhancing radiation sensitivity. Genes ChromosomesCancer 2003;38:330–8.

38. Jain RK. Normalization of tumor vasculature: anemerging concept in antiangiogenic therapy. Science2005;307:58–62.

39. Padera TP, Stoll BR, Tooredman JB, Capen D,di Tomaso E, Jain RK. Pathology: cancer cells compressintratumour vessels. Nature 2004;19:427–695.

40. Tong RT, Boucher Y, Kozin SV, Winkler F, Hicklin DJ,Jain RK. Vascular normalization by vascular endothelialgrowth factor receptor 2 blockade induces a pressuregradient across the vasculature and improves drugpenetration in tumors. Cancer Res 2004;64:3731–6.

41. Winkler F, Kozin SV, Tong RT, et al. Kinetics ofvascular normalization by VEGFR2 blockade governsbrain tumor response to radiation: role of oxygenation,angiopoietin-1, and matrix metalloproteinases. CancerCell 2004;6:553–63.

42. Abdollahi A, Lipson KE, Sckell A, et al. Combinedtherapy with direct and indirect angiogenesis inhibitionresults in enhanced antiangiogenic and antitumoreffects. Cancer Res 2003;63:8890–8.

43. Abdollahi A, Domhan S, Jenne JW, et al. Apoptosissignals in lymphoblasts induced by focused ultrasound.FASEB J 2004;18:1413–4.

44. Bischof M, Abdollahi A, Gong P, et al. Triplecombination of irradiation, chemotherapy (peme-trexed), and VEGFR inhibition (SU5416) in humanendothelial and tumor cells. Int J Radiat Oncol BiolPhys 2004;60:1220–32.

45. Nagel S, Wagner S, Koziol J, Kluge B, Heiland S.Volumetric evaluation of the ischemic lesion size withserial MRI in a transient MCAO model of the rat:comparison of DWI and T1WI. Brain Res Brain ResProtoc 2004;12:172–9.

46. Thali MJ, Dirnhofer R, Becker R, Oliver W, Potter K.Is ‘virtual histology’ the next step after the ‘virtualautopsy’? Magnetic resonance microscopy in forensicmedicine. Magn Reson Imaging 2004;22:1131–8.

47. Gupta AK, McKenna WG, Weber CN, et al. Localrecurrence in head and neck cancer: relationship toradiation resistance and signal transduction. ClinCancer Res 2002;8:885–92.

48. Abdollahi A, Li M, Ping G, et al. Inhibition of platelet-derived growth factor signaling attenuates pulmonaryfibrosis. J Ex Med. In press 2005.

49. Garcia-Barros M, Paris F, Cordon-Cardo C, et al.Tumor response to radiotherapy regulated by endothe-lial cell apoptosis. Science 2003;300:1155–9.

50. Klement G, Baruchel S, Rak J, et al. Continuous low-dose therapy with vinblastine and VEGF receptor-2antibody induces sustained tumor regression withoutovert toxicity. J Clin Invest 2000;105:R15–24.

51. Pietras K, Rubin K, Sjoblom T, et al. Inhibition ofPDGF receptor signaling in tumor stroma enhancesantitumor effect of chemotherapy. Cancer Res 2002;62:5476–84.

52. Pietras K, Ostman A, Sjoquist M, et al. Inhibition ofplatelet-derived growth factor receptors reduces inter-stitial hypertension and increases trans-capillary trans-port in tumors. Cancer Res 2001;61:2929–34.

53. Bergers G, Song S, Meyer-Morse N, Bergsland E,Hanahan D. Benefits of targeting both pericytes andendothelial cells in the tumor vasculature with kinaseinhibitors. J Clin Invest 2003;111:1287–95.

54. Griffin RJ, Williams BW, Wild R, Cherrington JM, ParkH, Song CW. Simultaneous inhibition of the receptorkinase activity of vascular endothelial, fibroblast, andplatelet-derived growth factors suppresses tumorgrowth and enhances tumor radiation response. CancerRes 2002;62:1702–6.

55. Pietras K, Hanahan D. A Multitargeted, Metronomic,and Maximum-tolerated dose ‘‘chemo-switch’’ regimenis antiangiogenic, producing objective responses andsurvival benefit in a mouse model of cancer. J ClinOncol. Epub ahead of print, 2004 Dec 21.

56. Franke TF, Yang SI, Chan TO, et al. The proteinkinase encoded by the Akt proto-oncogene is a target ofthe PDGF-activated phosphatidylinositol 3-kinase. Cell1995;81:727–36.

Beneficial Effects of a Trimodal Anticancer Strategy

www.aacrjournals.org 3655 Cancer Res 2005; 65: (9). May 1, 2005

Research. on February 25, 2021. © 2005 American Association for Cancercancerres.aacrjournals.org Downloaded from

Page 14: Trimodal Cancer Treatment: Beneficial Effects of Combined ... · Trimodal Cancer Treatment: Beneficial Effects of Combined Antiangiogenesis, Radiation, and Chemotherapy Peter E. Huber,

2005;65:3643-3655. Cancer Res   Peter E. Huber, Marc Bischof, Jürgen Jenne, et al.   Antiangiogenesis, Radiation, and ChemotherapyTrimodal Cancer Treatment: Beneficial Effects of Combined

  Updated version

  http://cancerres.aacrjournals.org/content/65/9/3643

Access the most recent version of this article at:

   

   

  Cited articles

  http://cancerres.aacrjournals.org/content/65/9/3643.full#ref-list-1

This article cites 51 articles, 26 of which you can access for free at:

  Citing articles

  http://cancerres.aacrjournals.org/content/65/9/3643.full#related-urls

This article has been cited by 24 HighWire-hosted articles. Access the articles at:

   

  E-mail alerts related to this article or journal.Sign up to receive free email-alerts

  Subscriptions

Reprints and

  [email protected] at

To order reprints of this article or to subscribe to the journal, contact the AACR Publications

  Permissions

  Rightslink site. (CCC)Click on "Request Permissions" which will take you to the Copyright Clearance Center's

.http://cancerres.aacrjournals.org/content/65/9/3643To request permission to re-use all or part of this article, use this link

Research. on February 25, 2021. © 2005 American Association for Cancercancerres.aacrjournals.org Downloaded from