Review Radiation Cemo

download Review Radiation Cemo

of 15

Transcript of Review Radiation Cemo

  • 8/2/2019 Review Radiation Cemo

    1/15

    REVIEW

    www.nature.com/clinicalpractice/onc

    The concurrent chemoradiation paradigmgeneral principlesTanguy Y Seiwert*, Joseph K Salama and Everett E Vokes

    INTRODUCTION

    Three clinical rationales support the use ofchemotherapy delivered concurrently withradiation. First, concomitant chemoradio-therapy can be used with organ-preservingintent, resulting in improved cosmesis andfunction compared with surgical resection withor without adjuvant treatment. Second, chemo-therapy can act as a radiosensitizer, improving

    the probability of local control and, in somecases, survival, by aiding the destruction of radi-oresistant clones (Table 1). Third, chemotherapygiven as part of concurrent chemoradiationmay act systemically and potentially eradicatedistant micrometastases.

    CLINICAL APPLICATIONDISEASE

    ENTITIES

    Currently, concomitant chemoradiotherapy iswidely used in the treatment of solid tumors.In almost all malignancies in which loco-regional control is necessary, concurrent chemo-radiotherapy is either an established treatmentmodality or is actively being investigated.The optimum schedules, synergistic combi-nation of agents, and integration of targetedtherapies are also areas of active investigation.Chemoradiotherapy combinations for individualdiseases with their specific indications and limita-tions are beyond the scope of this article; however,an overview of the most common uses is shownin Table 1. In the next issue of this journal, theapplication of chemoradiotherapy in head andneck cancer will be detailed, exemplifying some

    of the principles outlined here.

    THEORETICAL FRAMEWORK FOR

    THERAPYTHE STEEL PARADIGM

    In 1979, Steel and Peckham introduced a theo-retical framework to describe the interaction ofcytotoxic chemotherapy and radiotherapy.1 Theterm spatial cooperation is used to describethe scenario whereby radiotherapy acts loco-regionally, and chemotherapy acts against distantmicrometastases, without interaction between

    During the past 20 years, the advent of neoadjuvant, primary, and adjuvantconcurrent chemoradiotherapy has improved cancer care dramatically.Significant contributions have been made by technological improvementsin radiotherapy, as well as by the introduction of novel chemotherapyagents and dosing schedules. This article will review the rationale for theuse of concurrent chemoradiotherapy for treating malignancies. Themolecular basis and mechanisms of action of combining classic cytotoxicagents (e.g. platinum-containing drugs, taxanes, etc.) and novel agents

    (e.g. tirapazamine, EGFR inhibitors and other targeted agents) withradiotherapy will be examined. This article is part one of two articles.In the subsequent article, the general principles outlined here will beapplied to head and neck cancer, in which the impact of concurrentchemoradiotherapy is particularly evident.

    KEYWORDS chemoradiation, cytotoxic, radiation, resistance, synergistic

    TY Seiwert is a fellow in the Department of Medicine, University of ChicagoCancer Research Center, JK Salama is an instructor in the Departmentof Radiation and Cellular Oncology, Pritzker School of Medicine, TheUniversity of Chicago, and EE Vokes is the John E Ultmann Professorof Medicine and Radiation and Cellular Oncology, and Director of theHematology/Oncology Section in the Department of Medicine, PritzkerSchool of Medicine, The University of Chicago, IL, USA.

    Correspondence*University of Chicago, 5841 South Maryland Avenue, MC 2115, Chicago, IL 606371470, USA

    [email protected]

    Received 20 March 2006 Accepted 18 September 2006

    www.nature.com/clinicalpractice

    doi:10.1038/ncponc0714

    REVIEW CRITERIAThe information for this Review was compiled using the PubMed andMEDLINE databases for articles published until 15 June 2006. Electronic early-release publications were also included. Only articles published in Englishwere considered. The search terms used included chemoradiotherapy orchemoradiation in association with the following search terms: reviews,radiosensitizer, concurrent, mechanism, molecular, cell cycle, cytotoxicchemotherapy, hypoxia, targeted therapies, radioresistance, cisplatin,tirapazamine, carboplatin, oxaliplatin, 5-FU, gemcitabine, capecitabine,pemetrexed, paclitaxel, mitomycin C, hydroxyurea, temozolomide,amifostine, palifermin, EGFR, Met, STAT 1, and VEGF. Full articles wereobtained and references were checked for additional material when appropriate.References were chosen based on the best clinical or laboratory evidence, especiallyif the work had been corroborated by published work from other centers. Prioritywas given to studies in high impact factor journals when available.

    SUMMARY

    86 NATURE CLINICAL PRACTICE ONCOLOGY FEBRUARY 2007 VOL 4 NO 2

  • 8/2/2019 Review Radiation Cemo

    2/15

    REVIEW

    FEBRUARY 2007 VOL 4 NO 2 SEIWERT ET AL. NATURE CLINICAL PRACTICE ONCOLOGY 87

    www.nature.com/clinicalpractice/onc

    the agents (Figure 1). This cooperative effectrequires the agents to have non-overlappingtoxicity profiles in order that both modali-ties can be used at effective doses withoutincreasing normal tissue effects. When combinedconcurrently with radiotherapy, however, fewchemotherapeutic agents meet this criterion,because limited single-agent activity or toxicity-driven dose reductions preclude the deliveryof systemic dosing schedules. Many trials of

    concomitant chemoradiotherapy, however, havedemonstrated decreased incidence of distantmetastases compared with radiation alone. Thisevidence could indicate that chemotherapy deliv-ered at radiosensitizing doses has some systemicspatial cooperative effect, or that the improvedlocal control of chemoradiotherapy decreasessubsequent metastases.

    The second interaction between radiation andchemotherapy is radiation sensitization, which iseither additive or supra-additive: the interaction

    within the radiation field leads to increasedkilling of cells (cytotoxic activity) either to thesame degree as (additive) or more than (supra-additive) using both modalities sequentially(Figures 1 and 2).Strictly speaking, radiosensi-tizers shouldnt have inherent cytotoxic activity.The hypoxic cell sensitizers (e.g. misonidazole),and thymidine analogs (e.g. bromodeoxyuridine)are examples of true radiosensitizers; however,the radiosensitizers most commonly used today

    (cisplatin, 5-fluorouracil [5-FU], and taxanes) dohave inherent cytotoxic activity and can increasedamage to normal tissues, with a true benefitachieved only if the increase in antitumor effect islarger than the normal tissue damage (Figure 3).

    Infra-additivedrugs possess radioprotectiveproperties that lessen the cytoxic effect of radia-tion on the tumor and/or normal tissue. Ideally,such agents should be selective for normaltissue to allow administration of higher radia-tion doses. Several agents are being investigated

    Table 1 Overview of disease entities and indications in which concurrent chemoradiotherapy is used.a

    Disease entity Indication and treatment Commonly used agents Benefit

    Upper aerodigestive tract cancers

    Head and neck cancer Locally advanced HNCprimary or adjuvant treatment

    Cisplatin, 5-FU, FHX,cetuximab

    Improved organ preservation and survivalcompared with radiation alone

    Non-small-cell lung cancer Stage IIIB, nonoperablenonmetastatic disease Cisplatin, carboplatin/paclitaxel, cisplatin/etoposide Curative approach in poor surgicalcandidates or IIIB disease

    Small-cell lung cancer Limited stage disease Cisplatin/etoposide Curative in ~20% of patients

    Esophageal cancer Locally advanced disease Cisplatin/5-FU Survival benefit, increased cure rates,organ preservation

    Gastrointestinal malignancies

    Rectal cancer Neoadjuvant 5-FU Improved sphincter preservation, decreasein local and distal failures

    Anal cancer Mainstay of curative treatment 5-FU, MMC Improved organ preservation

    Gastric cancer Adjuvant Cisplatin, 5-FU Some data indicate a survival benefit

    Pancreatic cancer Adjuvant, unresectablelocoregionally advanced tumors

    5-FU Improved locoregional control, possibly asurvival benefit

    Cholangiocarcinoma Adjuvant, unresectablelocoregionally advanced tumors

    5-FU Some data indicate a survival benefit

    Gynecological and genitourinary cancers

    Cervical cancer Primary modality Cisplatin, 5-FU, hydroxyurea Improved local and distal control,organ preservation

    Bladder cancer Primary modality Cisplatin Improved local control

    Other cancers

    Glioblastoma Adjuvant Temozolomide Survival benefit

    Sarcoma Neoadjuvant Doxorubicin Downstaging, improved organ preservation

    aThis is a limited overview, and concurrent chemoradiotherapy is used in most solid tumors either as a standard treatment or investigationally. For further details pleaserefer to the organ-specific literature. Abbreviations: 5-FU, 5-fluorouracil; FHX, 5-FU, hydroxyurea and radiation; HNC, head and neck cancer; MMC, mitomycin C.

  • 8/2/2019 Review Radiation Cemo

    3/15

    REVIEW

    88 NATURE CLINICAL PRACTICE ONCOLOGY SEIWERT ET AL. FEBRUARY 2007 VOL 4 NO 2

    www.nature.com/clinicalpractice/onc

    and have shown promising early results withradiation alone, but for concurrent chemo-radiotherapy definitive phase III evidence doesnot currently support their routine use. One suchagent, amifostinean organic thiophosphatemight decrease cisplatin-induced or radiation-induced toxicity by acting as a scavenger of freeradicals, as well as by binding to and neutral-izing organoplatinums and alkylating agents,thereby preventing DNA adduct formation.Although decreased incidence of xerostomiain patients with head and neck cancer treatedwith radiotherapy alone was convincinglydemonstrated in a large trial,2 the benefit withconcurrent chemoradiotherapy remains unclear,as demonstrated by a phase III study showing nosignificant difference in xerostomia or mucositisrates.3 Although a single-institution trial of

    patients with advanced lung cancer randomizedto receive concurrent cisplatin, etoposide, andtwice-daily radiation demonstrated a decreasein grade 12 esophagitis, grade 3 pneumonitis,and neutropenic fever, an increase in sneezing,dysgeusia (loss of taste), and more importantlyhypotension, was observed.4 In a larger phaseIII trial conducted by the Radiation TherapyOncology Group, addition of amifostine toradiotherapy did not demonstrate any differ-ence in grade 3 or greater esophagitis, although

    patients had a statistically significant, althoughsmall (1.3%) decrease in weight loss.3 Concernsthat radiation protectors might have a tumorprotective effect has limited their use; however,two large meta-analyses did not confirm thisconcern.5,6 At doses lower than those used for

    normal tissue protection, amifostine has beenshown to have antimutagenic properties.7The agent palifermin, a recombinant human

    keratinocyte growth factor, reduces oralmucositis in patients undergoing radiation andchemotherapy for autologous stem-cell trans-plant and in metastatic colorectal cancer treatedwith 5-FU.8,9 This drug is currently undergoingphase III testing with concurrent chemoradio-therapy in patients with advanced head andneck cancer.

    Determination of additive effects between

    chemotherapy and radiation

    The type of interaction between chemotherapyand radiotherapy within the radiation field(supra-additivity, additivity, or infra-additivity)can be determined. For this purpose, Steel andPeckham described the isobologram analysis,which is based on an isoeffect concept forchemoradiotherapy interaction (Figure 2).1Independent doseresponse curves for chemo-therapy and radiotherapy are necessary to createa plot, called an isobologram. The isobologramis generated by plotting the dose of each agent(i.e. chemotherapy and radiotherapy) againsteach other, which produces a cytotoxic effect(isoeffect) on the axes of increasing dose of eachagent. Two curves, named mode 1 and mode 2,can then be generated (Figure 2). The mode 1curve results from the assumption that radia-tion and chemotherapy act independently, andis created by plotting a given dose of radiationagainst the dose of chemotherapy needed toproduce an effect equal to the difference betweenthe chosen cytotoxic effect and the effect of thecurrent dose of radiation. The mode 2 curve

    assumes that radiation and chemotherapy haveidentical mechanisms of action. Points on thiscurve are generated by plotting doses of radia-tion against the doses of chemotherapy needed toincrease the effect of the dose of radiation to thechosen cytotoxic effect. Multiple points on bothof these lines are obtained by varying the radia-tion dose and calculating the appropriate dosesof chemotherapy. Finally, the true (empiric)survival curve is generated for radiation andchemotherapy given in combination. The doses

    No interaction

    modalities work

    independently

    Chemotherapy

    toxicities

    Radiation

    toxicities

    Molecular level

    Cellular level

    Tissue level

    Independent toxicities

    Radiosensitization

    Spatial cooperation In-field cooperation

    Infra-additivity

    (antagonism)a

    Locoregional

    control

    AdditivitySupra-additivity

    (synergism)

    Chemotherapy:

    distal control(out-of-field)

    Radiation:

    local control(in-field)

    Synergistic toxicities

    Figure 1 Rationale for adding chemotherapy to radiation. Spatial and in-fieldcooperation are the two idealized types of cooperation between radiation and

    chemotherapy. Both mechanisms can contribute synergistically to clinical

    benefit.aUsually not desirable as this could protect the tumor.

  • 8/2/2019 Review Radiation Cemo

    4/15

    REVIEW

    FEBRUARY 2007 VOL 4 NO 2 SEIWERT ET AL. NATURE CLINICAL PRACTICE ONCOLOGY 89

    www.nature.com/clinicalpractice/onc

    of each agent needed to produce the chosen cyto-toxic effect are plotted. Points that fall below theenvelope between mode 1 and 2 curves indicatesupra-additivity, data points occurring within theenvelope (between the two curves) indicate addi-tivity, and data points above the envelope indi-

    cate infra-additivity. Further details and samplecalculations are provided elsewhere.10,11Other methods exist to determine types of

    additivity, including the median effect prin-ciple and the response surface approach, bothof which are described in detail elsewhere.1215Unfortunately, the concept of additivity is oflimited use in clinical practice, as preclinicalprediction of additivity does not translate wellinto clinical outcomes. The use of this methodis, therefore, limited to forming hypotheses,which need to be confirmed empirically.

    Quantification of the chemotherapy

    and radiation interaction

    The radiosensitizing ability of a drug can beexpressed by the therapeutic ratio. This ratio isderived from sigmoid-shaped doseresponsecurves, calculated by plotting the response oftissues (both normal and tumor) on the ordi-nate axis versus the chemotherapy or radiationtherapy dose on the abscissa (Figure 3). Thetherapeutic ratio is defined as the quotient ofthe dose that produces a 50% tumor control rateand the dose that produces a 50% normal tissuetoxicity rate. When chemotherapy is combinedwith radiation, both normal tissue and tumorcontrol curves produced by radiation aloneshift to the left because of the chemotherapy-induced sensitization of cells. Ideally, radiationsensitizers should influence the tumor responsecurve more than the normal tissue curve, thusresulting in a greater than one therapeutic ratio.Similar to the concept of additivity, therapeuticratios are mainly hypothesis-forming and needto be tested empirically in phase I and II studies.Such preclinical data may help in the initial dose

    schedule selection for phase I trials.

    Mechanisms of radiation resistance or failure

    Tumors have developed multiple strategies toresist radiation damage. Table 2 gives an over-view of the most widely accepted mechanisms.Simplistically, larger tumors have a stochasticchance that some cells will survive radiotherapy.Moreover, hypoxic tumor cells have increasedresistance to radiation; multiple studies havedemonstrated that hypoxic cells exhibit 2.53.0

    times the resistance to radiotherapy damagecompared with normoxic cells.1620 As shown inFigure 4, the phases of the cell cycle significantlyinfluence the radiosensitivity of cells.21

    1.0

    0.5

    0.0

    0.0 0.5

    Drug dose

    RTdose

    1.0

    Supra-additivity

    (synergism)

    Infra-additivity(antagonistic effect)

    Additivity envelope(area between the

    mode 1 [upper] andmode 2 [lower] curves)

    Figure 2Schematic example of an isobologram depicting the combinationof radiation and a systemic agent. Thex andyaxes show the isoeffective

    levels for radiation and drug. The thick line is the line of additivity, and the

    additivity envelope is based on the combined standard errors. Curves above

    the envelope represent antagonistic effects and curves below the envelope

    represent synergistic effects.1,41 Abbreviation: RT, radiotherapy.

    100

    0

    Radiation dose (Gy)

    Response/toxicityrat

    e(%)

    Normal tissue

    Tumor

    Figure 3Schematic doseresponse curves for tumor and normal tissuedamage with radiation. The offset between the two curves indicates the

    therapeutic range. Chemoradiotherapy leads to a shift of both curves to the

    left, ideally with a stronger shift of the tumor curve (as indicated by the longer

    arrow), increasing overall efficacy of treatment (radiation enhancement).120

  • 8/2/2019 Review Radiation Cemo

    5/15

    REVIEW

    90 NATURE CLINICAL PRACTICE ONCOLOGY SEIWERT ET AL. FEBRUARY 2007 VOL 4 NO 2

    www.nature.com/clinicalpractice/onc

    One of the most commonly invoked under-lying mechanisms in treatment failure22,23 istumor cell repopulation, which results in a rapidgrowth between radiation fractions. Althoughincompletely understood, the stimulation ofgrowth factors and the selection of resistantclones lead to rapid emergence of treatmentresistance.22,24,25 In addition, rapidly prolif-erating tumors contain a high proportion ofradioresistant cells in the S phase of the cellcycle. For further information on repopulation,readers should refer to the excellent review bySchmidt-Ullrich et al.26

    Certain tumors are intrinsically radio-

    resistant, while others acquire mechanismsof radioresistance during treatment. Intrinsicradioresistance is seen when there is anincreased surviving fraction of tumor cells after2 Gy radiotherapy; this result is often depictedin a clonogenic radiation cell survival curveassay. Small increases in radioresistance leadto large, logarithmic decreases in final cell killafter radiotherapy. In some tumors, such evalu-ation in radioresistance in pretreatment biop-sies predicts for radiosensitivity and clinical

    outcome, although these findings have so farnot been widely reproducible.27,28 Activationof certain prosurvival pathways that preventapoptosis can induce treatment resistance.Among the many pathways implicated aremutated p53,29 amplification of DNA repairgenes, increased levels of reactive oxygen speciesscavengers, and activation of prosurvival/poor-prognosis oncogenes such as EGFR,30,31 orc-MET(also known as MST1R).32,33 Recently,gene-expression profiling has identifiedthat radiation-resistant tumors overexpressmany genes related to interferon pathways orinduced by interferon itself, especiallySTAT1.

    Radiosensitive cells transfected with STAT1demonstrated radioprotection after exposureto 3.0 Gy radiation.34

    General mechanisms of chemotherapy

    and radiotherapy interaction

    Seven major interactions between chemotherapyand radiation will be explained here and are listedin Table 3. For most chemotherapeutic agentsseveral interactions apply at the same time.First, DNA damage can be induced by both

    Table 2 Mechanisms of radioresistance.

    Process affected Mechanism Comments

    Large tumor cellburden

    Tumor size is inversely correlated with tumor response.Radiation-induced cell kill is a random eventthe higherthe number of cells, the higher the chance of cellsescaping a lethal hit.121,122

    Upfront or completion surgery should be considered toreduce tumor bulk or residual disease.

    Tumor cellmicroenvironment/hypoxia

    Oxygen is needed to generate ROS and other radicals withradiation. ROS are thought to be essential to the cytotoxiceffect from radiation (reviewed in Cook et al.123).

    Hypoxia is present for two reasons:

    1. increased interstitial pressure may causehypoperfusion, hypoxia and acidosis;124126

    2. cancer-related anemia contributes to local hypoxia(HIF1 is a marker of tumor hypoxia).

    Hypoxic cells are 2.53.0 times less radiation-sensitive thannormoxic cells.18,44

    Both hypoxia and HIF1 are adverse prognostic factors.127

    Chemotherapy can increase radiation effect:

    1. through reoxygenation second to tumor shrinkage(e.g. with paclitaxel46);

    2. by killing hypoxic cells selectively (e.g. withtirapazamine or mitomycin C);

    3. through resensitization of hypoxic cells to radiation(nitroimidazolesin development).

    Inherent oracquired tumorcell resistance

    Multiple mechanisms are thought to contribute, includingmutated p5329, DNA repair gene amplification, increasedlevels of ROS scavengers, activation of prosurvival/poor-prognosis oncogenes (EGFR,100,101

    c-MET32).

    Delays or interruptions in radiotherapy are known to leadto the development of radioresistance and allow suchresistant cells to repopulate.

    Repopulation Regrowth of tumor cells between doses of radiotherapyor chemotherapy. Accelerated repopulation might lead totreatment failure and emergence of true radioresistance(see row above).22

    Accelerated radiation schemes are intended to preventrepopulation.128

    Antimetabolites with activity in the S phase of the cell cycle(5-FU, hydroxyurea) also inhibit repopulation.

    EGFR inhibitors can block cell proliferation betweenradiotherapy fractions.101

    Abbreviations: 5-FU, 5-fluorouracil; HIF1, hypoxia-inducible factor 1-alpha; ROS, reactive oxygen species.

  • 8/2/2019 Review Radiation Cemo

    6/15

    REVIEW

    FEBRUARY 2007 VOL 4 NO 2 SEIWERT ET AL. NATURE CLINICAL PRACTICE ONCOLOGY 91

    www.nature.com/clinicalpractice/onc

    chemotherapy and radiotherapy and synergy ispossible. Ionizing radiation induces DNA basedamage, alkali-labile sites, single-strand breaks, anddouble-strand breaks (DSBs). All of these errorscan be rapidly repaired except for DSBs, which ifnot repaired are considered lethal.35,36 The integra-tion of cisplatin into DNA or RNA in close prox-imity to a radiation-induced single-strand breakcan act synergistically to make the defect signifi-cantly more difficult to repair (Figure 5).37,38Second, chemotherapy can inhibit post-radiationdamage repair. DNA synthesis and repair sharecommon pathways, which provide the rationalefor using DNA synthesis inhibitors with radiationas a means of inducing cytotoxic damage to tumorcells. Agents affecting nucleoside and nucleotidemetabolism can inhibit the repair of radiation-induced DNA damage in patients, and are amongthe most potent radiation sensitizers. Examples of

    such agents include the fluoropyrimidines,thymidine analogs, gemcitabine, and hydroxyurea.Third, administered concurrently, radiotherapyand chemotherapy often target different phases ofthe cell cycle and may cooperate to produce anadditive effect (i.e. cytokinetic cooperation/synchronization). The radiosensitivity of a cell isdependent on the phase of the cell cycle; cells inthe S phase are the most radioresistant, and cellsin the G2M phase of the cell cycle are the mostradiosensitive (Figure 4).39,40 In addition,

    cytokinetic cooperation of S-phase-specific agents(e.g. camptothecins, 5-FU, hydroxyurea) is seen ifcells are exposed in close temporal proximityto radiation.41 Some drugs (e.g. taxanes) are able

    to synchronize with the cell cycle of tumor cellsto allow increased efficacy of subsequent radio-therapy (called synchronization or cell-cyclepooling). This process was successfully shownin vitro, although the conceptespecially itsapplicability in vivoremains controver-sional.11,42,43 Fourth, the increased resistance ofhypoxic cells to radiation (Table 2) means thathypoxic cell sensitizers may be beneficial.18,44

    Hypoxia is common in many cancers andwas shown to be a marker of aggressive clinicalbehavior and poor prognosis.45 Chemotherapycan help eliminate these resistant cells andincrease the efficacy of radiotherapy viamultiple mechanisms (Table 3). Tirapazamine,and potentially mitomycin C, preferentiallykill hypoxic cells. Additionally, paclitaxel,46and EGFR inhibitors,24 were shown to shrinktumors, thereby increasing perfusion andoxygenation and reducing radioresistant hypoxicareas. This hypoxic effect might be applicablefor many other agents. Fifth, repopulation ofrapidly proliferating tumors is usually mediatedby overexpression of growth factors and growthfactor receptors, as well as increased activity of

    downstream signaling pathways, and the pres-ence of activating mutations in genes involved atall levels of the signaling pathways. Agents thattarget the S phase of the cell cycle, such as 5-FU,irinotecan, and hydroxyurea, as well as those thatinhibit proliferation and/or growth factor path-ways, such as EGFR inhibitors, may be effectivein preventing tumor cell repopulation, therebyradiosensitizing tumor cells.24,47

    In addition to their antiproliferative effectsthat prevent tumor cell repopulation, EGFR

    Most radiosensitive

    Most radioresistant

    Division

    Taxanes

    Vinca alkaloids

    Etoposide

    Vinca alkaloids

    5-fluorouracil

    Methotrexate

    Hydroxyurea Doxorubicin

    Cytarabine

    Gemcitabine

    Etoposide

    Cell-cycle-independent: Platinating agents

    Alkylating agents

    G2

    M

    S

    G0

    G1 Bleomycin

    Figure 4 Cell-cycle schematic and respective

    sensitivity to chemotherapeutic agents.

    Cell deathRepairRepair

    Radiation-induced

    single-strand breakaCisplatin adduct Cisplatin and radiation-

    induced damage

    Figure 5 Increased DNA damage by addition of cisplatin to radiation.aRadiation can also induce other DNA damage, of which double-strand breaks

    are considered lethal.

  • 8/2/2019 Review Radiation Cemo

    7/15

    REVIEW

    92 NATURE CLINICAL PRACTICE ONCOLOGY SEIWERT ET AL. FEBRUARY 2007 VOL 4 NO 2

    www.nature.com/clinicalpractice/onc

    inhibitors and antiangiogenic agents can blocksignaling pathways that are responsible for

    aggressive tumor biology, poor prognosis, andradioresistance. Although the exact mecha-nisms of various targeted therapies vary andare generally poorly understood, preclinical48and clinical49 data support the rationale forradiosensitizing properties as a consequenceof inhibiting the detrimental effects of theagents targets. Finally, some tumors resistantto standard chemoradiotherapy respond toalterations in radiation fractionation. Thisphenomenon, termed hyperradiation sensitivity,

    is observed at radiation doses greater than 1 Gy.Preclinically, for both paclitaxel and docetaxel,

    low-dose fractionated radiation can overcomeradioresistance.50 Clinical trials evaluating thistechnique are currently underway.

    Specific mechanisms of chemotherapy

    and radiotherapy interaction

    Platinum analogsCisplatin is one of the most commonly useddrugs for concurrent chemoradiotherapy.Through interactions with nucleophilic sites onDNA and RNA, cisplatin introduces intrastrand

    Table 3 Mechanisms of chemotherapy and radiotherapy interaction.

    Process affected Mechanisma Drug examples

    Increased radiationdamagea

    Incorporation of chemotherapy drug into DNA/RNA 5-FU: incorporation into DNA, increasing susceptibilityto RT damage

    Cisplatin: cross-links with DNA or RNA (intrastrand andinterstrand); works for both hypoxic and oxygenated

    cells51

    Inhibition of DNA repairprocessa

    Interference with the DNA repair process afterradiation

    Halogenated pyrimidines (e.g. 5-FU, bromodeoxyuridine,iododeoxyuridine)

    Nucleoside analogs (e.g. gemcitabine, fludarabine)

    Cisplatin

    Methotrexate

    Camptothecins and doxorubicin

    Etoposide

    Hydroxyurea

    Carmustine, lomustine

    Cell-cycle interference(cytokinetic cooperationand synchronization)a

    Most cytotoxic chemotherapies as well as radiationare cell-cycle-specific, and proliferating cells are mostsusceptible

    Accumulation of cells in the G2 and M phases(the most radiosensitive phases)

    Elimination of radioresistant cells in the S phase

    Taxanes lead to cell-cycle arrest via tubulin stabilization

    Nucleoside analogs (e.g. gemcitabine, fludarabine),etoposide, methotrexate, hydroxyurea

    Enhanced activityagainst hypoxic cellsa

    Reoxygenation second to tumor shrinkage. Hypoxiccells are 2.53.0 times less radiation-sensitive thannormoxic cells18,44

    Chemotherapy can help to eliminate hypoxic cells

    Most chemotherapeutic agents; described in particularfor paclitaxel45

    Tirapazamine, mitomycin (selective killing of hypoxic cells);nitroimidazoles (resensitize hypoxic cells to radiation)

    Radiotherapyenhancement bypreventing repopulationa

    Systemic therapy can slow or stop rapid proliferation,which could otherwise be the basis for repopulationphenomenon

    Most chemotherapeutic agents, in particular:

    Antimetabolites with activity in the S phase inhibitrepopulation (e.g. 5-FU, hydroxyurea)

    EGFR inhibitors, which impede cell proliferation betweenRT fractions100

    Inhibition of prosurvival

    and poor prognosismarkersa

    Targeted therapies (best demonstrated for EGFR

    inhibition) block signaling pathways that might beresponsible for radioresistance and poor prognosis

    EGFR inhibitorsshown for anti-EGFR antibody, PKI-

    166 (small-molecule TKI), and EGFR antisense,129131

    but on the basis of clinical experience likely to be a classeffect49,132

    Hyperradiationsensitivityb

    HNSCC cells resistant to standard-fraction CRT canbe resensitized to CRT by using smaller fraction sizes(

  • 8/2/2019 Review Radiation Cemo

    8/15

    REVIEW

    FEBRUARY 2007 VOL 4 NO 2 SEIWERT ET AL. NATURE CLINICAL PRACTICE ONCOLOGY 93

    www.nature.com/clinicalpractice/onc

    and interstrand cross-links, thereby distorting theDNA structure, and blocking nucleotide replica-tion and transcription. Active in both hypoxicand well-oxygenated cells,51 several potentialmechanisms for cisplatin-mediated radiationsensitization were reported and summarized by

    Wilson and co-workers.41

    It has been proposedthat radiation induces free radicals and subse-quently the formation of toxic platinum inter-mediates, which increase cell killing.52 Moreover,ionizing radiation can increase cellular uptakeof platinum.53 Damage to DNA by ionizingradiation that typically would be repairable canbecome fixed and lethal through cisplatins free-electron-scavenging capacity. This inhibition ofDNA repair (Figure 5)54 leads to an increasedincidence of cell-cycle arrest and apoptotic celldeath after radiation.41,55

    In vitro studies have shown that the mostsynergistic combination of cisplatin and radia-tion involves low doses of each,either of whichwould be insufficient to cause cell death if admin-istered alone. Cisplatin would seem to inhibitthe sublethal damage repair process implicatedin the recovery of insufficiently radiated cells.41Radiosensitization by cisplatin and carboplatinmay be limited to cells with an intact homo-logous recombination repair system,41,56 butradiosensitization is not impacted by hypoxia.Oxaliplatin, a cisplatin-related compound,has been shown to have activity in cisplatin-resistant systems and unaltered sensitivity inmutated mismatch-repair systems.49 Althoughless well studied, oxaliplatin is postulated tohave similar radiosensitization mechanismsto those of cisplatin.41

    Antimetabolite-based chemoradiotherapy

    5-fluorouracilThe halogenated pyrimidine nucleoside analog5-FU is used extensively with radiation.57,58 Thisanalog impedes nucleic acid synthesis throughthymidylate synthase inhibition, depleting the

    pool of nucleotide triphosphates, leading to cell-cycle changes, DNA fragmentation, and ultimatelycell death.59 When 5-FU is incorporated into RNAand DNA it inhibits not only DNA synthesis, butalso transcription and protein synthesis. Optimal5-FU radiosensitization requires continuousadministration of the agent during radiationbecause of the need for continuous thymidy-late synthase inhibition, the short half-life ofplasma 5-FU and intracellular phosphorylated5-FU metabolites.

    5-FU radiosensitization is postulated to occurby the agents effect on the proportion of cells inthe radioresistant S phase of the cell cycle.Whengiven in standard doses, 5-FU is able to kill cellsin the S phase. Additionally, at sublethal concen-trations, 5-FU pre-incubation is hypothesized to

    radiosensitize tumor cells through changes inthe S-phase cell-cycle checkpoint, which allowinappropriate progression out of the S phaseinto the G2 phase. This conjecture is supportedby the fact that blocking the entry of cells intoS phase prevents radiosensitization for a similarpyrimidine analog, fluorodeoxyuridine.57,58Impaired repair of radiation-induced DSBsmight also contribute to 5-FU cytotoxicity andradiosensitization.57,58

    Capecitabine

    Capecitabine is an oral prodrug that is convertedto 5-FU via thymidine phosphorylase. Radiationhas been shown to preferentially increase tumorthymidine phosphorylase levels via induction oftumor necrosis factor. In a colon cancer fluoro-pyrimidine-resistant xenograft model, exposureto capecitabine before irradiation demonstrateda supra-additive effect compared with radiationalone.60 Clinical trials are actively exploring therole of the combination of capecitabine andradiation therapy.

    GemcitabineGemcitabine is a widely used S-phase cell-cycle-specific pyrimidine analog that hinders DNAsynthesis and repair through depletion of deoxy-nucleoside triphosphates that are required by twoenzymes: DNA polymerase and ribonucleotidereductase. Gemcitabine has shown activity inmany solid tumors, either alone or in combina-tion with other agents (e.g. cisplatin or carbo-platin).61 Initial clinical studies of pancreaticand lung cancer demonstrated marked toxicityof gemcitabine-based chemoradiotherapy anddampened enthusiasm for the efficacy of this

    drug, since only doses much lower than thoseused without radiation could be administeredsafely. Recent data from multi-institutionalstudies have shown that full doses of gemcit-abine can be delivered with aggressive conformalradiation techniques.6264

    In preclinical models, the potent radio-sensitizing properties of gemcitabine weremost pronounced with exposure to low dosesof the agent at least 24 h before irradiation.Interestingly, sensitization persisted for up to

  • 8/2/2019 Review Radiation Cemo

    9/15

    REVIEW

    94 NATURE CLINICAL PRACTICE ONCOLOGY SEIWERT ET AL. FEBRUARY 2007 VOL 4 NO 2

    www.nature.com/clinicalpractice/onc

    48 h, and radiation exposure before exposureto gemcitabine did not lead to sensitization.These observations are consistent with the timeneeded to deplete deoxynucleoside triphosphates(dATP) and transition through the S phase ofthe cell cycle.6567 It seems that incorporation

    of incorrect bases secondary to deoxynucleosidetriphosphate depletion and S-phase accumulationare the basis of the radiosensitizing propertiesof gemcitabine.65

    PemetrexedThis compound is a novel multitargeted antifolatethat inhibits thymidylate synthase, dihydrofolatereductase, and glycinamide ribonucleotideformyl-transferase, all of which are key enzymesinvolved in nucleotide synthesis. Preclinical datasuggest synergistic antitumor activity of peme-

    trexed and concurrent radiotherapy, and inter-ference with DNA synthesis is thought to be theprimary cytotoxic mechanism.68 Pemetrexedis not cell-cycle-specific as it shows equal effi-cacy in the G1 and S phases of the cell cycle.69Although the exact interaction of pemetrexedwith radiation is not fully understood, it hasbeen postulated that prolongation of the S phaseby radiation increases the intracellular drugexposure time and toxicity. Pemetrexed-basedchemoradiotherapy is in phase I clinical testingin several tumor types.70

    HydroxyureaHydroxyurea acts as a radiation enhancer in vitroand in vivo51 and is useful for the treatment ofhead and neck cancer.71 This drug was previouslyused for squamous-cell carcinoma of the cervix(replaced by cisplatin)72 and gliomas (replacedby temozolomide),73 and was proposed for thetreatment of pancreatic cancer (replaced by 5-FUand potentially gemcitabine).74 Ribonucleotide-reductase inhibition prevents radiation-inducedDNA damage repair during nucleotide excision.Furthermore, hydroxyurea might also synchronize

    cancer cells at the G1S checkpoint.74 The doubleaction of hydroxyurea of cell-cycle synchronizingand DNA damage repair inhibition has beensuggested as a mechanism for its more efficaciousaction as a radiation sensitizer at doses lowerthan for the ribonucleotide-reductase inhibitorsgemcitabine and trimidox.74 Antimetabolitessuch as hydroxyurea are selectively cytotoxicto cells that are in the relatively radioresistantS phase of the cell cycle, which might contributeto overcoming radioresistance.

    Taxane-based chemoradiotherapy

    Paclitaxel and docetaxel form high-affinity bondswith microtubules, promoting tubulin poly-merization and stabilization. At high doses, thesedrugs block prophase to metaphase progression,disrupting the centrosome network and thereby

    causing cell death.75

    Despite the structural simi-larities of these two agents, differences in excre-tion and cell-cycle tropism instigate differingtemporal interactions of paclitaxel and docetaxelwith radiation.76

    Two mechanisms for paclitaxel and docetaxelradiosensitization have been proposed. Thestandard explanation is that cells remain inthe G2M phase of the cell cycle, leadingto synchronization (cell-cycle pooling) oftumor cells at a point of maximum radio-sensitivity,11,42 but it is still unclear whether

    cell-cycle redistribution occurs, especiallyin vivo.43True synchronization in large tumorsseems unlikely, and may not increase the thera-peutic index as normal cells are also affected.77Alternatively, taxanes can induce tumorshrinkage, improving perfusion and subsequentreoxygenation. As hypoxic areas of the tumorare reoxygenated they become more sensitiveto radiation-induced cell kill.46 Paradoxically,low doses of taxanes may induce protectionagainst radiation via possible alterations insignal transduction pathways. Pretreatmentof a human laryngeal squamous cell line with7.5 nmol/l paclitaxel 6 h before irradiationinduced subadditive effects via G2 blockade.78Extensive single-agent and chemoradiotherapytaxane mechanisms are reviewed by Hennequinand Favaudon.11

    Mitomycin-C-based chemoradiotherapy

    Mitomycin C inhibits DNA and RNA synthesisby interfering with DNA cross-linking, prima-rily at the guanine and cytosine pairs. Althoughmitomycin C is not cell-cycle-specific, this agentis known to induce marked cell-cycle arrest at the

    G2M transition.79,80 In combination with radia-tion, mitomycin C acts as a hypoxic cell sensitizerand is thought to prevent repopulation, althoughthe exact mechanism remains elusive.81

    Tirapazamine-based chemoradiotherapy

    Tirapazamine is the lead compound in a class thathas selective cytotoxic activity under hypoxic con-ditions (hypoxic cell cytotoxic).82 Hypoxic tumorcells that are relatively resistant to radiotherapyexhibit aggressive growth behavior, and portend

  • 8/2/2019 Review Radiation Cemo

    10/15

    REVIEW

    FEBRUARY 2007 VOL 4 NO 2 SEIWERT ET AL. NATURE CLINICAL PRACTICE ONCOLOGY 95

    www.nature.com/clinicalpractice/onc

    a poor prognosis. Although widely recognized inits ability to enhance radiation,83 debate existsas to whether tirapazamines effects are addi-tive or supra-additive.84,85 Regardless, theor-etical models show that the killing of hypoxiccells might improve outcomes compared with

    standard radiosensitizers.86

    Tirapazamines approximately 100-foldincreased potency under anoxic conditionsoccurs via electron donation, which causesformation of transient oxidizing radicals. Innormoxic tissue these radicals quickly bindavailable molecular oxygen, re-establishingthe nontoxic parent compound. In the absenceof oxygen, however, these oxidizing radicalsinduce the formation of DNA radicals byextracting a proton from the C4 location of thedeoxyribose ring on the DNA.87 This process

    can lead to cytotoxic DNA-strand breaks.Additionally, unknown mechanisms decreasetopoisomerase II activity in tirapazamine-treated cells.88 Although little or no cell killingis observed in normoxic cells, systemic sideeffects including fatigue, muscle cramps, andreversible ototoxicity were observed during theclinical administration of tirapazamine, andhave been attributed to a loss of mitochondrialmembrane potential.89

    Temozolomide-based chemoradiotherapy

    Temozolomide is an orally administered cyto-toxic alkylating agent, which readily crosses thebloodbrain barrier; 30% of plasma concentra-tions are achieved in the cerebrospinal fluid.90Commonly used to treat gliomas, temozolo-mide causes DNA damage by methylationof the O-6 position of guanine and activatesthe p53-controlled DNA damage responsepathway.91 Tumors with methylation of theO-6-methylguanine DNA-methyltransferase(MGMT), a p53 DNA damage repair enzyme,are preferentially radiosensitized.92,93 Temo-zolomide also inhibits signaling of radiation-

    triggered cell migration and invasiveness94 anddecreases tumor cell repopulation.

    The aim of combining temozolomide withradiation is the use of an intrinsically activeagent that has a different toxicity profile toradiation.95In a study by Wedge et al.,96 a glio-blastoma cell line with no MGMT activity wascompared with a colorectal cancer cell line withhigh repair activity. Temozolomide and radiationwere additive in the glioblastoma line, whereasantagonism was observed in the colorectal

    cancer line. This finding is probably attributableto radiation induction of MGMT. Additionally,temozolomide and radiation showed additiveand supra-additive activity.97

    Advances in radiotherapy

    While most advances in concurrent chemoradio-therapy have focused on the integration of novelsystemic agents, recent technological improve-ments in radiotherapy have impacted directlyon concurrent chemoradiotherapy. Intensity-modulated radiation therapy (IMRT) has allowedbetter delivery of radiation to the target volumes,allowing sharp dose gradients between targetsand normal tissues. After clinical implementa-tion of IMRT, decreased acute gastrointestinal,skin, hematologic, and salivary toxicity rateswere reported, which improved the therapeutic

    index of radiation.

    98

    These decreases in normaltissue toxicity could also improve the therapeuticindex of chemoradiation. Additionally, imple-mentation of image-guided radiotherapy vialinear accelerator-based kilovoltage techniques,and gating radiation delivery with the respira-tory cycle, will improve day-to-day targeting andpotentially decrease acute and chronic toxicitiesof chemoradiotherapy.Molecular-targeted therapies in

    combination with chemoradiotherapy

    In the past decade, the promises of molecular-targeted agents have increasingly come to frui-tion, and these agents have been combined withradiotherapy in an effort to optimize the thera-peutic index of drug dosing. Molecular-targetedtherapies are an attractive option combined withchemoradiotherapy because they are more specificfor the target and can inhibit radioresistance path-ways. An extensive review of molecular-targetedagents and their interaction with radiotherapy hasbeen published by Ma et al.99

    EGFR-targeted therapies and chemoradiotherapy

    The membrane-bound receptor tyrosine kinaseEGFR is activated upon binding of the ligand(transforming growth factor , epidermalgrowth factor), which induces dimerizationand subsequent phosphorylation of the intra-cellular EGFR tyrosine residues. Upon activation,intracellular signaling cascades mediate variouscellular responses important for tumor survivaland growthnamely, increased proliferation,invasion, angiogenesis, and metastasis, andconcomitant decreased apoptosis.

  • 8/2/2019 Review Radiation Cemo

    11/15

    REVIEW

    96 NATURE CLINICAL PRACTICE ONCOLOGY SEIWERT ET AL. FEBRUARY 2007 VOL 4 NO 2

    www.nature.com/clinicalpractice/onc

    EGFR (erbB1) and the EGFR family memberserbB24 are deregulated in head and neck,lung, breast, and colorectal cancers. IncreasedEGFR protein expression correlates withincreased tumor size, recurrence risk, and radio-resistance, and with decreased survival.100,101

    Cell culture experiments of radiation-inducedEGFR expression and increased radioresistance,demonstrated by the addition of exogenousEGF, indicated a causal link between EGFR andradioresistance.102

    Preclinical studies with the EGFR inhibi-tors cetuximab, gefitinib, and erlotinib showenhanced radiosensitivity leading to supra-additive efficacy both in vitro and in vivo.49,100,103Proposed mechanisms for radiosensitiza-tion via EGFR inhibitors include inhibitionof cell proliferation, impairment of DNA

    damage repair,

    104

    attenuation of tumor neo-angiogenesis, inhibition of radiation-inducedEGFR nuclear import,105 and promotion ofradiation-induced apoptosis.106,107 In partic-ular, the antiproliferative effects of EGFR inhi-bition most likely prevent repopulation,100 amajor mechanism implicated in radioresistance.The other mechanisms may well have a role inthe supra-additivity of the anti-EGFR radiationcombination.

    Antiangiogenic and anti-VEGF therapyin combination with radiationAngiogenesis is essential for sustained tumorgrowth, and many new cancer therapies aredirected against modification of the tumorvasculature.108,109 The process of angiogenesisis mediated by multiple proangiogenic andantiangiogenic factors, with VEGF having acentral role.108,109 Anti-VEGF agents fall intotwo broad categories: those that target the VEGRligand, such as bevacizumab, and those thattarget the receptor, such as PTK787 (an antibodyto VEGFR-2). Other antiangiogenic strategiesinclude antiangiogenic factor administration to

    counterbalance proangiogenic stimuli, inhibitionof extracellular matrix degradation enzymes,integrin antagonists, and therapies with directendothelial cell toxicity.

    Two mechanisms for radiosensitization withantiangiogenic agents have been proposed andcould exist in parallel.109,110 The traditionalview is that antiangiogenic destruction oftumor vessels leads to hypoxia and starvation,which paradoxically could also increase tumorresistance.111 Increasingly, however, findings

    show that increases in blood flow, oxygen, anddrug delivery (i.e. a transient normalization ofthe abnormal structure and function of tumorvessels) is seen as the underlying mechanism ofantiangiogenic therapies.109

    Radiosensitizing properties were first

    reported for angiostatin when Weichselbaumand colleagues demonstrated that the combina-tion of angiostatin and radiation was synergisticand decreased radioresistance.112 These resultshave also been confirmed for antiangiogenicsmall-molecule tyrosine kinase inhibitors.113Additionally, endostatin showed additivity withradiation regarding tumor regression, growthinhibition, angiogenesis, and enhanced apop-tosis.114 These mechanisms could be caused byenhancement of tumor oxygenation, leading toincreased radiosensitivity, as well as direct tumor

    growth delay.

    110,112

    Novel targeted therapies with radiosensitizingpropertiesReceptor tyrosine kinases other than EGFR arealso potential targets for novel radiosensitizers.Overexpression of c-Met has been linked to poorprognosis in many cancers,115 and synergismwith radiation has been described in glioblastomacell lines.116 Other receptor tyrosine kinases suchas insulin-like growth factor receptor or ephrinreceptors are additional candidates. Anotherinnovative strategy that has been tested in phase Iclinical trials is radiation-induced activation ofgene transcription. For example, TNFerade(GenVec, Gaithersburg, MD) is a replication-deficient adenovirus vector with an early growthresponse protein 1 (EGR1) radiation-induciblepromoter leading to intratumoral human tumornecrosis factor production. Initial applications ofthis agent in esophageal, rectal, and pancreaticcancer, as well as in sarcomas, have shown proofof principal, with extensive tumor necrosis as asign of activity.117 Additional studies in head andneck cancer are ongoing.

    With the increasing understanding of cancerbiology, hundreds of novel targeted agents arescheduled to come into preclinical and clinicaldevelopment in the next decade. Among theseagents proteasome inhibitors, and agents thattarget mammalian target of rapamycin, haveshown promising results when combined withradiotherapy.118,119 For further information onthe interaction of targeted agents with radiation,readers are referred to the excellent review by Maand coauthors.99

  • 8/2/2019 Review Radiation Cemo

    12/15

    REVIEW

    FEBRUARY 2007 VOL 4 NO 2 SEIWERT ET AL. NATURE CLINICAL PRACTICE ONCOLOGY 97

    www.nature.com/clinicalpractice/onc

    CONCLUSION

    In summary, concurrent chemoradiotherapy is ahighly efficacious locoregional treatment optionfor solid tumors, which can be used alone orcombined with surgery or induction/consolidationchemotherapy. Through our increased under-

    standing of the molecular basis of chemotherapyand radiation interactions, we have gained insightsinto how to best use and potentially combineagents with radiation, with benefits beginningto be seen in patients. Novel targeted therapiesand agents specific for hypoxic or radiated cellshold promise for further significant improve-ment of therapeutic ratios. Concurrent chemo-radiotherapy already offers excellent locoregionalcontrol with an acceptable toxicity profile for thetreatment of many locoregional advanced tumors.In the final article of this two-article series we will

    examineusing the example of head and neckcancerspecific concurrent chemoradiotherapytreatment options and the underlying clinicalevidence. Head and neck cancer is often a loco-regionally confined disease, and this is exemplifiedby the profound impact and benefit that concurrentchemoradiotherapy can have for patients.

    KEY POINTS

    Concurrent chemoradiotherapy has improved

    cancer care during the past two decades in

    multiple diseases, and can be used in the

    neoadjuvant, primary (definitive), or adjuvant

    setting

    Chemotherapy or targeted agents can increase

    the efficacy of radiation

    Radiosensitizing effects (interaction within the

    radiation field) can be additive or supra-additive

    Multiple mechanisms underlie radiosensitizing

    properties of chemotherapeutic agents and

    include increased radiation damage, inhibition

    of DNA repair, cell-cycle synchronization,

    increased cytotoxicity against hypoxic cells,

    inhibition of prosurvival pathways, and

    abrogation of rapid tumor cell repopulation

    Radioresistance occurs through multiple

    mechanisms, such as a large tumor burden,

    hypoxia, rapid tumor cell repopulation, as well

    as the constitutive or acquired activation of

    radioresistance signaling pathways

    In addition to the classic chemotherapeutic

    agents with radiosensitizing properties (i.e.

    cisplatin and paclitaxel), several novel agents

    show promising interactions with radiation (e.g.

    EGFR inhibitors, pemetrexed, tirapazamine, and

    potentially several other targeted therapies)

    References

    1 Steel GG and Peckham MJ (1979) Exploitable

    mechanisms in combined radiotherapy-

    chemotherapy: the concept of additivity. Int J Radiat

    Oncol Biol Phys5: 8591

    2 Brizel DM et al. (2000) Phase III randomized trial of

    amifostine as a radioprotector in head and neck

    cancer.J Clin Oncol18: 33393345

    3 Buentzel J et al. (2006) Intravenous amifostine during

    chemoradiotherapy for head-and-neck cancer:

    a randomized placebo-controlled phase III study.

    Int J Radiat Oncol Biol Phys64: 684691

    4 Komaki R et al. (2004) Effects of amifostine

    on acute toxicity from concurrent chemotherapy

    and radiotherapy for inoperable non-small-cell lung

    cancer: report of a randomized comparative trial.

    Int J Radiat Oncol Biol Phys58: 13691377

    5 Malik R et al. (2005) Meta-analysis of the effect of

    amifostine on response rates in advanced non-small

    cell lung cancer patients treated on randomized trials.

    Int J Radiat Oncol Biol Phys63 (Suppl 1): S117

    6 Sasse AD et al. (2006) Amifostine reduces side

    effects and improves complete response rate during

    radiotherapy: results of a meta-analysis. Int J Radiat

    Oncol Biol Phys64: 784791

    7 Grdina DJ et al. (2002) Radioprotectants: currentstatus and new directions. Oncology63 (Suppl 2):

    S2S10

    8 Stiff PJ et al. (2006) Palifermin reduces patient-

    reported mouth and throat soreness and improves

    patient functioning in the hematopoietic stem-cell

    transplantation setting.J Clin Oncol [doi: 10.1200/

    JCO.2005.02.8340]

    9 Rosen LS et al. (2006) Palifermin reduces the

    incidence of oral mucositis in patients with metastatic

    colorectal cancer treated with fluorouracil-based

    chemotherapy.J Clin Oncol24: 51945200

    10 Giocanti N et al. (1993) DNA repair and cell cycle

    interactions in radiation sensitization by the

    topoisomerase II poison etoposide. Cancer Res53:

    21052111

    11 Hennequin C and Favaudon V (2002) Biological basis

    for chemo-radiotherapy interactions. Eur J Cancer38:223230

    12 Chou TC and Talalay P (1984) Quantitative analysis

    of dose-effect relationships: the combined effects

    of multiple drugs or enzyme inhibitors.Adv Enzyme

    Regul22: 2755

    13 Reynolds CP and Maurer BJ (2005) Evaluating

    response to antineoplastic drug combinations in tissue

    culture models. Methods Mol Med110: 173183

    14 Teicher BA (2003) Assays for in vitro andin vivo

    synergy. Methods Mol Med85: 297321

    15 White DB et al. (2003) A new nonlinear mixture

    response surface paradigm for the study of

    synergism: a three drug example. Curr Drug Metab4:

    399409

    16 Coleman CN (1988) Hypoxia in tumors: a paradigm

    for the approach to biochemical and physiologicheterogeneity.J Natl Cancer Inst80: 310317

    17 Gray LH et al. (1953) The concentration of oxygen

    dissolved in tissues at the time of irradiation as a

    factor in radiotherapy. Br J Radiol26: 638648

    18 Kumar P (2000) Tumor hypoxia and anemia: impact

    on the efficacy of radiation therapy. Semin Hematol

    37: 48

    19 Overgaard J et al. (1989) Misonidazole combined with

    split-course radiotherapy in the treatment of invasive

    carcinoma of larynx and pharynx: report from the

    DAHANCA 2 study. Int J Radiat Oncol Biol Phys16:

    10651068

    20 Moulder JE and Rockwell S (1984) Hypoxic fractions

    of solid tumors: experimental techniques, methods

  • 8/2/2019 Review Radiation Cemo

    13/15

    REVIEW

    98 NATURE CLINICAL PRACTICE ONCOLOGY SEIWERT ET AL. FEBRUARY 2007 VOL 4 NO 2

    www.nature.com/clinicalpractice/onc

    of analysis, and a survey of existing data. Int J Radiat

    Oncol Biol Phys10: 695712

    21 Terasima T and Tolmach LJ (1963) X-ray sensitivity

    and DNA synthesis in synchronous populations of

    HeLa cells. Science140: 490492

    22 Fowler JF and Lindstrom MJ (1992) Loss of local

    control with prolongation in radiotherapy. Int J Radiat

    Oncol Biol Phys23: 457467

    23 Trott KR and Kummermehr J (1985) What is known

    about tumour proliferation rates to choose between

    accelerated fractionation or hyperfractionation?

    Radiother Oncol3: 19

    24 Krause M et al. (2005) Decreased repopulation

    as well as increased reoxygenation contribute to

    the improvement in local control after targeting of

    the EGFR by C225 during fractionated irradiation.

    Radiother Oncol76: 162167

    25 Grillo-Ruggieri F and Mantello G (2004) Is one Gy

    equal to one Gy after treatment interruptions? Rays

    29: 275278

    26 Schmidt-Ullrich RK et al. (1999) Molecular

    mechanisms of radiation-induced accelerated

    repopulation. Radiat Oncol Investig7: 321330

    27 West CM et al. (1997) The independence of intrinsic

    radiosensitivity as a prognostic factor for patient

    response to radiotherapy of carcinoma of the cervix.Br J Cancer76: 11841190

    28 Deacon J et al. (1984) The radioresponsiveness of

    human tumours and the initial slope of the cell survival

    curve. Radiother Oncol2: 317323

    29 Dey S et al. (2003) Low-dose fractionated radiation

    potentiates the effects of paclitaxel in wild-type and

    mutant p53 head and neck tumor cell lines. Clin

    Cancer Res9: 15571565

    30 Nix PA et al. (2004) Radioresistant laryngeal cancer:

    beyond the TNM stage. Clin Otolaryngol Allied Sci29:

    105114

    31 Smith BD and Haffty BG (1999) Molecular markers

    as prognostic factors for local recurrence and

    radioresistance in head and neck squamous cell

    carcinoma. Radiat Oncol Investig7: 125144

    32 Aebersold DM et al. (2001) Involvement of the

    hepatocyte growth factor/scatter factor receptor

    c-met and of Bcl-xL in the resistance of oropharyngeal

    cancer to ionizing radiation. Int J Cancer96: 4154

    33 Uchida D et al. (2001) Role of HGF/c-met system

    in invasion and metastasis of oral squamous cell

    carcinoma cellsin vitro and its clinical significance. Int

    J Cancer93: 489496

    34 Khodarev NN et al. (2004) STAT1 is overexpressed

    in tumors selected for radioresistance and confers

    protection from radiation in transduced sensitive cells.

    Proc Natl Acad Sci USA101: 17141719

    35 Bennett CB et al. (1993) Lethality induced by a single

    site-specific double-strand break in a dispensable

    yeast plasmid. Proc Natl Acad Sci USA15: 1317

    36 Ward JF (1998) Nature of lesions formed by ionising

    radiation. In DNA damage and repair: DNA repair

    in higher eukaryotes, 6584 (Eds Nickoloff JA andHoekstra MF). Totowa, NJ: Humana Press

    37 Begg AC (1990) Cisplatin and radiation: interaction

    probabilities and therapeutic possibilities. Int J Radiat

    Oncol Biol Phys19: 11831189

    38 Yang LX et al. (1995) Production of DNA double-

    strand breaks by interactions between carboplatin

    and radiation: a potential mechanism for

    radiopotentiation. Radiat Res143: 309315

    39 Sinclair WK and Morton RA (1966) X-ray sensitivity

    during the cell generation cycle of cultured Chinese

    hamster cells. Radiat Res29: 450474

    40 Terasima T and Tolmach LJ (1961) Changes in x-ray

    sensitivity of HeLa cells during the division cycle.

    Nature190: 12101211

    41 Wilson GD et al. (2006) Biologic basis for combining

    drugs with radiation. Semin Radiat Oncol16: 29

    42 Hei TK et al. (1994) Taxol and ionizing radiation:

    interaction and mechanisms. Int J Radiat Oncol Biol

    Phys29: 267271

    43 Stromberg JS et al. (1995) Lack of radiosensitization

    after paclitaxel treatment of three human carcinoma

    cell lines. Cancer75: 22622268

    44 Vaupel P (2004) Tumor microenvironmental

    physiology and its implications for radiation oncology.

    Semin Radiat Oncol14: 198206

    45 Semenza GL (2003) Targeting HIF-1 for cancer

    therapy. Nat Rev Cancer3: 721732

    46 Milas L et al. (1995) Tumor reoxygenation as a

    mechanism of taxol-induced enhancement of tumor

    radioresponse.Acta Oncol34: 409412

    47 Kim JJ and Tannock IF (2005) Repopulation of cancer

    cells during therapy: an important cause of treatment

    failure. Nat Rev Cancer5: 516525

    48 Liang K et al. (2003) The epidermal growth factor

    receptor mediates radioresistance. Int J Radiat Oncol

    Biol Phys57: 246254

    49 Bonner JA et al. (2006) Radiotherapy plus cetuximab

    for squamous-cell carcinoma of the head and neck.

    N Engl J Med354: 567578

    50 Spring PM et al. (2004) Low dose fractionatedradiation potentiates the effects of taxotere in nude

    mice xenografts of squamous cell carcinoma of head

    and neck. Cell Cycle3: 479485

    51 Vokes EE and Weichselbaum RR (1990) Concomitant

    chemoradiotherapy: rationale and clinical experience

    in patients with solid tumors.J Clin Oncol8: 911934

    52 Richmond RC (1984) Toxic variability and radiation

    sensitization by dichlorodiammineplatinum(II)

    complexes in Salmonella typhimurium cells. Radiat

    Res99: 596608

    53 Yang LX et al. (1995) Irradiation enhances cellular

    uptake of carboplatin. Int J Radiat Oncol Biol Phys33:

    641646

    54 Amorino GP et al. (1999) Radiopotentiation by the oral

    platinum agent, JM216: role of repair inhibition. Int J

    Radiat Oncol Biol Phys44: 399405

    55 Moreland NJ et al. (1999) Modulation of drug

    resistance mediated by loss of mismatch repair by

    the DNA polymerase inhibitor aphidicolin. Cancer Res

    59: 21022106

    56 Dolling JA et al. (1999) Cisplatin-modification of

    DNA repair and ionizing radiation lethality in yeast,

    Saccharomyces cerevisiae. Mutat Res433: 127136

    57 Lawrence TS et al. (1997) Fluoropyrimidine-radiation

    interactions in cells and tumors. Semin Radiat Oncol

    7: 260266

    58 McGinn CJ and Lawrence TS (2001) Recent

    advances in the use of radiosensitizing nucleosides.

    Semin Radiat Oncol11: 270280

    59 Pinedo HM and Peters GF (1988) Fluorouracil:

    biochemistry and pharmacology.J Clin Oncol6:

    16531664

    60 Sawada N et al. (1999) X-ray irradiation inducesthymidine phosphorylase and enhances the efficacy

    of capecitabine (Xeloda) in human cancer xenografts.

    Clin Cancer Res5: 29482953

    61 Hui YF and Reitz J (1997) Gemcitabine: a cytidine

    analogue active against solid tumors.Am J Health

    Syst Pharm54: 162170

    62 Crane CH et al. (2002) Is the therapeutic index

    better with gemcitabine-based chemoradiation than

    with 5-fluorouracil-based chemoradiation in locally

    advanced pancreatic cancer? Int J Radiat Oncol Biol

    Phys52: 12931302

    63 Blackstock AW et al. (2005) Initial pulmonary toxicity

    evaluation of chemoradiotherapy (CRT) utilizing 74 Gy

    3-dimensional (3-D) thoracic radiation in stage III

  • 8/2/2019 Review Radiation Cemo

    14/15

    REVIEW

    FEBRUARY 2007 VOL 4 NO 2 SEIWERT ET AL. NATURE CLINICAL PRACTICE ONCOLOGY 99

    www.nature.com/clinicalpractice/onc

    non-small cell lung cancer (NSCLC): a Cancer and

    Leukemia Group B (CALGB) randomized phase II trial

    [abstract #7060]. Presented at the 2005 ASCO annual

    meeting, Orlando, FL

    64 Talamonti MS et al. (2006) A multi-institutional phase

    II trial of preoperative full-dose gemcitabine and

    concurrent radiation for patients with potentially

    resectable pancreatic carcinoma.Ann Surg Oncol13:

    150158

    65 Lawrence TS et al. (2003) The mechanism of action of

    radiosensitization of conventional chemotherapeutic

    agents. Semin Radiat Oncol13: 1321

    66 Lawrence TS et al. (1997) Delayed radiosensitization

    of human colon carcinoma cells after a brief exposure

    to 2',2'-difluoro-2'-deoxycytidine (Gemcitabine). Clin

    Cancer Res3: 777782

    67 Shewach DS and Lawrence TS (1996) Gemcitabine

    and radiosensitization in human tumor cells. Invest

    New Drugs14: 257263

    68 Bischof M et al. (2002) Interaction of pemetrexed

    disodium (ALIMTA, multitargeted antifolate) and

    irradiationin vitro. Int J Radiat Oncol Biol Phys52:

    13811388

    69 Bischof M et al. (2003) Radiosensitization by

    pemetrexed of human colon carcinoma cells in

    different cell cycle phases. Int J Radiat Oncol BiolPhys57: 289292

    70 Seiwert TY et al. (2005) A phase I dose-escalating

    study of combination pemetrexed-based

    chemotherapy and concomitant radiotherapy for

    locally advanced or metastatic non-small cell lung or

    esophageal cancer [abstract #7062]. Presented at the

    2005 ASCO annual meeting, Orlando, FL

    71 Vokes EE et al. (1992) Hydroxyurea with concomitant

    radiotherapy for locally advanced head and neck

    cancer. Semin Oncol19: 5358

    72 Kuo ML et al. (1997) The interaction of hydroxyurea

    and ionizing radiation in human cervical carcinoma

    cells. Cancer J Sci Am3: 163173

    73 Prados MD et al. (1998) Radiation therapy and

    hydroxyurea followed by the combination of 6-

    thioguanine and BCNU for the treatment of primary

    malignant brain tumors. Int J Radiat Oncol Biol Phys

    40: 5763

    74 Leyden D et al. (2000) Hydroxyurea and trimidox

    enhance the radiation effect in human pancreatic

    adenocarcinoma cells.Anticancer Res20: 133138

    75 Schiff PB and Horwitz SB (1980) Taxol stabilizes

    microtubules in mouse fibroblast cells. Proc Natl

    Acad Sci USA77: 15611565

    76 Hennequin C et al. (1995) S-phase specificity of cell

    killing by docetaxel (Taxotere) in synchronised HeLa

    cells. Br J Cancer71: 11941198

    77 Steel GG (1994) Cell synchronization unfortunately

    may not benefit cancer therapy. Radiother Oncol32:

    9597

    78 Ingram ML and Redpath JL (1997) Subadditive

    interaction of radiation and Taxolin vitro. Int J Radiat

    Oncol Biol Phys37: 1139114479 Heinrich MC et al. (1998) DNA cross-linker-

    induced G2/M arrest in group C Fanconi anemia

    lymphoblasts reflects normal checkpoint function.

    Blood91: 275287

    80 Sugiyama K et al. (2000) UCN-01 selectively

    enhances mitomycin C cytotoxicity in p53 defective

    cells which is mediated through S and/or G(2)

    checkpoint abrogation. Int J Cancer85: 703709

    81 Budach W et al. (2002) Mitomycin C in combination

    with radiotherapy as a potent inhibitor of tumour cell

    repopulation in a human squamous cell carcinoma.

    Br J Cancer86: 470476

    82 Wouters BG et al. (1999) Tirapazamine: a new drug

    producing tumor specific enhancement of platinum-

    based chemotherapy in non-small-cell lung cancer.

    Ann Oncol10 (Suppl 5): S29S33

    83 Brown JM and Lemmon MJ (1990) Potentiation by the

    hypoxic cytotoxin SR 4233 of cell killing produced by

    fractionated irradiation of mouse tumors. Cancer Res

    50: 77457749

    84 Lartigau E and Guichard M (1995) Does tirapazamine

    (SR-4233) have any cytotoxic or sensitizing effect on

    three human tumour cell lines at clinically relevant

    partial oxygen pressure? Int J Radiat Biol67: 211216

    85 Lambin P et al. (1992) The effect of the hypoxic cell

    drug SR-4233 alone or combined with the ionizing

    radiations on two human tumor cell lines having

    different radiosensitivity. Radiother Oncol24: 201204

    86 Brown JM and Koong A (1991) Therapeutic

    advantage of hypoxic cells in tumors: a theoretical

    study.J Natl Cancer Inst83: 178185

    87 Brown JM (1993) SR 4233 (tirapazamine): a new

    anticancer drug exploiting hypoxia in solid tumours.

    Br J Cancer67: 11631170

    88 Brown JM (1999) The hypoxic cell: a target for

    selective cancer therapyeighteenth Bruce F Cain

    Memorial Award lecture. Cancer Res59: 58635870

    89 Wouters BG et al. (2001) Mitochondrial dysfunction

    after aerobic exposure to the hypoxic cytotoxin

    tirapazamine. Cancer Res61: 14515290 Patel M et al. (2003) Plasma and cerebrospinal fluid

    pharmacokinetics of intravenous temozolomide in

    non-human primates.J Neurooncol61: 203207

    91 Hickman MJ and Samson LD (1999) Role of DNA

    mismatch repair and p53 in signaling induction of

    apoptosis by alkylating agents. Proc Natl Acad Sci

    USA96: 1076410769

    92 Roos W et al. (2004) Apoptosis triggered by DNA

    damage O6-methylguanine in human lymphocytes

    requires DNA replication and is mediated by p53 and

    Fas/CD95/Apo-1.Oncogene23: 359367

    93 Hermisson M et al. (2006) O6-methylguanine

    DNA methyltransferase and p53 status predict

    temozolomide sensitivity in human malignant glioma

    cells.J Neurochem96: 766776

    94 Wick W et al. (2002) Prevention of irradiation-induced

    glioma cell invasion by temozolomide involves

    caspase 3 activity and cleavage of focal adhesion

    kinase. Cancer Res62: 19151919

    95 Beauchesne P (2002) Promising survival and

    concomitant radiation plus temozolomide followed by

    adjuvant temozolomide.J Clin Oncol20: 31803181

    96 Wedge SR et al. (1997) In vitro evaluation of

    temozolomide combined with X-irradiation.

    Anticancer Drugs8: 9297

    97 van Rijn J et al. (2000) Survival of human glioma cells

    treated with various combination of temozolomide

    and X-rays. Int J Radiat Oncol Biol Phys47: 779784

    98 Salama JK (2004) Intensity-modulated radiation

    therapy in gynecologic malignancies. Curr Treat

    Options Oncol5: 97108

    99 Ma BB et al. (2003) Combined-modality treatment

    of solid tumors using radiotherapy and moleculartargeted agents.J Clin Oncol21: 27602776

    100 Ang KK et al. (2004) Epidermal growth factor receptor

    and response of head-and-neck carcinoma to

    therapy. Int J Radiat Oncol Biol Phys58: 959965

    101 Pal SK and Pegram M (2005) Epidermal growth factor

    receptor and signal transduction: potential targets for

    anti-cancer therapy.Anticancer Drugs16: 483494

    102 Dent P et al. (1999) Radiation-induced release of

    transforming growth factor alpha activates the

    epidermal growth factor receptor and mitogen-

    activated protein kinase pathway in carcinoma cells,

    leading to increased proliferation and protection

    from radiation-induced cell death. Mol Biol Cell10:

    24932506

  • 8/2/2019 Review Radiation Cemo

    15/15

    REVIEW

    www.nature.com/clinicalpractice/onc

    103 Chinnaiyan P et al. (2005) Mechanisms of enhanced

    radiation response following epidermal growth factor

    receptor signaling inhibition by erlotinib (Tarceva).

    Cancer Res65: 33283335

    104 Bandyopadhyay D (1998) Physical interaction

    between epidermal growth factor receptor and DNA-

    dependent protein kinase in mammalian cells.J Biol

    Chem273: 15681573

    105 Dittmann KH et al. (2001) Characterization of the amino

    acids essential for the photo- and radioprotective

    effects of a Bowman-Birk protease inhibitor-derived

    nonapeptide. Protein Eng14: 157160

    106 Milas Let al. (2000) In vivo enhancement of tumor

    radioresponse by C225 antiepidermal growth factor

    receptor antibody. Clin Cancer Res6: 701708

    107 Bonner JA et al. (2000) Enhanced apoptosis with

    combination C225/radiation treatment serves as the

    impetus for clinical investigation in head and neck

    cancers.J Clin Oncol18 (Suppl): 47S53S

    108 Folkman J (2006) Angiogenesis.Annu Rev Med57:

    118

    109 Jain RK (2005) Normalization of tumor vasculature: an

    emerging concept in antiangiogenic therapy. Science

    307: 5862

    110 Wachsberger P et al. (2003) Tumor response to

    ionizing radiation combined with antiangiogenesis orvascular targeting agents: exploring mechanisms of

    interaction. Clin Cancer Res9: 19571971

    111 Murata Ret al. (1997) An antiangiogenic agent

    (TNP-470) inhibited reoxygenation during fractionated

    radiotherapy of murine mammary carcinoma. Int J

    Radiat Oncol Biol Phys37: 11071113

    112 Mauceri HJ et al. (1998) Combined effects of

    angiostatin and ionizing radiation in antitumour

    therapy. Nature394: 287291

    113 Ning S et al. (2002) The antiangiogenic agents

    SU5416 and SU6668 increase the antitumor effects

    of fractionated irradiation. Radiat Res157: 4551

    114 Hanna NNet al. (2000) Antitumor interaction

    of short-course endostatin and ionizing radiation.

    Cancer J6: 287293

    115 Birchmeier C et al. (2003) Met, metastasis, motility

    and more. Nat Rev Mol Cell Biol4: 915925

    116 Lal B et al. (2005) Targeting the c-Met pathway

    potentiates glioblastoma responses to gamma-

    radiation. Clin Cancer Res11: 44794486

    117 Senzer N et al. (2005) Updated response and survival

    data for TNFerade combined with chemoradiation in

    the treatment of locally advanced pancreatic cancer

    (LAPC) [abstract #4097]. Presented at the 2005 ASCO

    annual meeting, Orlando, FL

    118 Russo SM et al. (2001) Enhancement of radiosensitivity

    by proteasome inhibition: implications for a role of

    NF-kappaB. Int J Radiat Oncol Biol Phys50: 183193

    119 Shinohara ET et al. (2005) Enhanced radiation

    damage of tumor vasculature by mTOR inhibitors.

    Oncogene24: 54145422

    120 Nishimura Y (2004) Rationale for chemoradiotherapy.

    Int J Clin Oncol9: 414420

    121 Pigott Ket al. (1995) Where exactly does failure occur

    after radiation in head and neck cancer? Radiother

    Oncol37: 1719

    122 Shukovsky LJ (1970) Dose, time, volume

    relationships in squamous cell carcinoma of the

    supraglottic larynx.Am J Roentgenol Radium Ther

    Nucl Med108: 2729

    123 Cook JAet al. (2004) Oxidative stress, redox, and the

    tumor microenvironment. Semin Radiat Oncol14:

    259266

    124 Milosevic M et al. (2004) The human tumor

    microenvironment: invasive (needle) measurement of

    oxygen and interstitial fluid pressure. Semin Radiat

    Oncol14: 249258

    125 Roh HD et al. (1991) Interstitial hypertension in

    carcinoma of uterine cervix in patients: possible

    correlation with tumor oxygenation and radiation

    response. Cancer Res51: 66956698

    126 Taghian AG et al. (2005) Paclitaxel decreases the

    interstitial fluid pressure and improves oxygenation in

    breast cancers in patients treated with neoadjuvantchemotherapy: clinical implications.J Clin Oncol23:

    19511961

    127 Koukourakis MI et al. (2002) Hypoxia-inducible

    factor (HIF1A and HIF2A), angiogenesis, and

    chemoradiotherapy outcome of squamous cell head-

    and-neck cancer. Int J Radiat Oncol Biol Phys53:

    11921202

    128 Suwinski R and Withers HR (2003) Time factor and

    treatment strategies in subclinical disease. Int J

    Radiat Biol79: 495502

    129 Holsinger FC et al. (2003) Epidermal growth factor

    receptor blockade potentiates apoptosis mediated by

    Paclitaxel and leads to prolonged survival in a murine

    model of oral cancer. Clin Cancer Res9: 31833189

    130 Knecht Ret al. (2003) EGFR antibody-supplemented

    TPE-chemotherapy: preclinical investigations to a

    novel approach for head and neck cancer induction

    treatment.Anticancer Res23: 47894795

    131 Niwa H et al. (2003) Antitumor effects of epidermal

    growth factor receptor antisense oligonucleotides

    in combination with docetaxel in squamous cell

    carcinoma of the head and neck. Clin Cancer Res9:

    50285035

    132 Cohen EE (2005) Integration of Gefitinib (G), into a

    Concurrent Chemoradiation (CRT) Regimen Followed

    by G Adjuvant Therapy in Patients with Locally

    Advanced Head and Neck Cancer (HNC)a Phase II

    Trial [abstract #5506].J Clin Oncol 23 (Suppl): 16S

    AcknowledgmentsThe authors would like to

    acknowledge the generous

    help of Dr Blase Polite

    and Dr Samir Undevia in

    reviewing organ-specific

    data.

    Competing interestsEE Vokes has declared

    associations with the

    following companies:

    AstraZeneca, Bristol-Myers

    Squibb, Eli Lilly, Genentech,

    ImClone, OSI and sanofi-

    aventis. See the article

    online for full details of

    the relationship. The other

    authors declared they have

    no competing interests.