22

Click here to load reader

Prevention of head and neck cancer: Current status and future prospects

Embed Size (px)

Citation preview

Page 1: Prevention of head and neck cancer: Current status and future prospects

agtlssacAgmtitcgisca

pwaSd

C0d

C

Prevention of Head and NeckCancer: Current Status and Future

Prospects

S urgery, radiation therapy, and chemotherapy have been the cor-nerstone of management of most neoplastic processes for almostall of the previous century. Although the former two modalities

re critical in providing local control of a disease, chemotherapy, whileenerally effective in providing systemic control, has been fraught withoxicities. Additionally, treatments with chemotherapeutic agents have aimited appeal due to their nonspecific mode of action. It is only in the lasteveral years that emphasis has been placed in identifying and targetingpecific molecular abnormalities in hematological malignancies, as wells solid tumors. There is now a better understanding of the biology ofell cycle, as well as perturbations that contribute to carcinogenesis.lthough in its infancy, developments in molecular biology andenetics have already made a significant impact in the management ofalignancies. Efforts in these disciplines are now beginning to come

o fruition, with almost daily reports of biological agents demonstrat-ng activity in several different tumors. It is well recognized, however,hat cancer is a multi-step, sustained disease process which begins as alonal abnormality. A previously normal cell must undergo a series ofenetic alterations, which culminate in a malignant phenotype. Thedentification of tamoxifen as an agent that prevents breast carcinogene-is, and of certain COX-2 inhibitors as effective preventive strategies inolonic polyp formation, has sparked interest in preventive, interruptivend reversal strategies in head and neck cancer as well.Squamous cell cancer of the head and neck (SCCHN) is a significantublic health problem worldwide, especially in India and Southeast Asia,here this disease accounts for 25% of all malignancies.1 Although there

re approximately 500,000 cases annually worldwide,1 in the Unitedtates alone, there are approximately 40,000 new cases, with 12,000

2,3

eaths annually. Even though significant advances have been made in

urr Probl Cancer 2004;28:265-286.147-0272/2004/$30.00 � 0oi:10.1016/j.currproblcancer.2004.05.003

urr Probl Cancer, September/October 2004 265

Page 2: Prevention of head and neck cancer: Current status and future prospects

tmnicsSwa

1hcnnsoshsr

MCIaeSiaisi

tmtuesi

2

he surgical, radiation therapy, and chemotherapeutic approaches to theanagement of this disease, the anatomical complexity of the head and

eck region almost invariably portends some degree of functionalmpairment. Despite adequate, definitive therapy, a well-acknowledgedonsequence of primary aerodigestive tract malignancies is the sustained,ignificant risk for the development of second primary tumors (SPTs).econd primary tumors of the upper aerodigestive tract and lung occurith significantly higher frequency in this patient population, at a rate of

pproximately 3 to 4% per year.3

The importance of SPTs in determining outcome was reported in the980s, when Vikram and coworkers4 observed that adequately treatedead and neck cancer patients continued to succumb to second malignan-ies. Similar findings have been reported by Cooper and coworkers,5 whooted that despite improvements in the local management of head andeck cancer, overall survival remained dismal due to the development ofecond malignancies. The above observations have been ratified by thosef Licciardello and coworkers.6 A review of the SEER database hashown a lifetime risk of SPTs of up to 25-40% in patients with a prioristory of laryngeal cancer.7,8 From a clinical and an epidemiologicaltandpoint, however, it is important to note that smoking cessation doesesult in a significant reduction in the incidence of SPTs.9-14

olecular Biology of Head and Neckarcinogenesis

nitially identified as activated cellular genes in certain malignancies, onlyfew members of this class have been directly implicated in tumorigen-

sis.15 Cyclin D1 overexpression has been specifically identified inCCHN, and its amplification results from abnormalities (amplification)

n chromosome 11q13. Up to 30% of SCCHN are associated withmplification of cyclin D1.16-18 A p53 analog (p40/51/63) has beendentified as a protooncogene and found to be frequently amplified inquamous cell cancers.19 Amplification at this locus correlates with bothncreased mRNA transcription and increased protein levels.20

Other, less well established perturbations in SCCHN include those inhe ras pathway. The eukaryotic transcription factor (IF4E) binds toRNA, and its overexpression results in upregulation of proteins essen-

ial for cell growth and division.21-25 Elevated protein levels can contrib-te to oncogenic transformation, as well as the stimulation of angiogen-sis through the elaboration of vascular endothelial growth factor.26 Aeparate, distinct class of gene products overexpressed in SCCHN

27

ncludes COX-2. Two separate isoforms of COX (1 and 2) exist.

66 Curr Probl Cancer, September/October 2004

Page 3: Prevention of head and neck cancer: Current status and future prospects

AguSaflIS

toSrcltutslOpaMepraat

itgiaiiS4

C

lthough COX-1 is constitutive and facilitates the synthesis of prosta-landins for several physiological functions, COX-2 is inducible and notsually detected in normal tissues. Its expression, however, is enhanced inCCHN28 from a variety of stimuli, including cytokines, growth factors,nd oncogenes.29-35 Interestingly, wild-type p53 suppresses COX-2unction, whereas mutations in p53 (detected commonly in precancerousesions as well as SCCHN36,37) result in a loss of this suppression.ncreased expression of COX-2 is seen in both precancerous lesions andCCHN.38-50

Analogous to gain in function with the acquisition of oncogene activity,he loss of critical tumor suppressor function is a well studied mechanismf tumorigenesis. The most commonly characterized abnormality inCCHN involves chromosome 9p21 to 22.51,52 Abnormalities in thisegion are also present in a high frequency in dysplasia as well asarcinoma in situ.53 The specific, well-characterized abnormality at thisocus consists of loss of p16 function, which normally serves to abrogatehe cyclin D1 pathway. Therefore, loss of p16 function facilitatesncontrolled cell proliferation. Although only 10 to 15% of head and neckumors were subsequently found to be associated with p16 abnormalitiespecifically,54,55 alternative mechanisms of p16 function inhibition wereater identified, implicating p16 in a significant proportion of SCCHN.ne such perturbation was identified in p16�, which resides upstream of16 on chromosome 9p21.56 This transcript encodes for a protein throughn alternate reading frame (ARF) which normally functions to bind toDM2, which in turn inhibits p53 degradation.57,58 As would be

xpected, introduction of p16 or p16� into SCCHN cell lines results inotent growth inhibition.59 Izzo and coworkers reported on the increasedisk for the development of invasive disease in lesions associated withltered cyclin D1 expression, during and after treatment of patients withbiochemopreventive approach consisting of 13-cis-retinoic acid, �-in-

erferon, and �-tocopherol.60

The p53 gene resides on chromosome 17p, which is frequently deletedn up to 60% of SCCHN.61 Interestingly, p53 mutations occur much laterhan 17p abnormalities, suggesting that an alternate tumor suppressorene, not yet identified, probably resides on chromosome 17 and is likelynvolved in early tumorigenesis. Shin and coworkers proposed that p53lterations occur early in tumorigenesis and may serve as an importantntermediate biomarker of chemopreventive efforts. They performedmmunohistochemical analyses for p53 expression in 33 patients withCCHN and identified altered expression in 45% of the malignant lesions,

5% of the dysplastic lesions, 29% of the hyperplastic lesions, 21% of the

urr Probl Cancer, September/October 2004 267

Page 4: Prevention of head and neck cancer: Current status and future prospects

nwspctshaSpanrpw1

aepvtntuiacsf

elpaippmb

2

ormal epithelium adjacent to the tumors, but in none of the controlsithout a diagnosis of SCCHN.62 Lippman and coworkers reported

imilarly on different levels of p53 expression in subjects with oralremalignant lesions.63 p53 expression was not altered in any of theontrols, but was notably altered in the deeper layers of the epithelium ofhe subjects with the premalignant lesions. Also, there was noted to be aignificant association between the level of p53 expression and theistologic grade. In a cohort of patients with SCCHN, p53 expression wasssociated with a worse outcome following definitive local therapy.64

hin and coworkers analyzed tumor specimens for p53 expression in 69atients with SCCHN treated with definitive surgery or radiation therapyt MD Anderson Cancer Center (MDACC) between 1980 and 1983. Theyoted that p53 expression was associated with an earlier tumor recurrenceate, as well as second primary tumor development in this patientopulation. Other loss of function abnormalities have also been associatedith chromosome 3p65,66 abnormalities, as well as loss of chromosome0q.67,68

The principal risk factors for the development of head and neck cancerre exposure to tobacco and alcohol. However, the majority of subjectsxposed to these carcinogens do not develop malignancies. There areresumably underlying genetic mechanisms that predispose certain indi-iduals to carcinogenesis. Genetic predisposition is further supported byhe observation that first-degree relatives of patients who have notecessarily been exposed to the carcinogens remain at an approximatelyhree- to eightfold higher risk of developing malignancies than thenrelated population.69-72 Advances in molecular biology now allow forsolation of genetic material (DNA) from the primary tumor for thessessment of specific molecular abnormalities (deletions/amplifications/haracterization of candidate oncogenes). Elucidation of the stepwiseequence of events in carcinogenesis now presents us with unique targetsor preventive, diagnostic, and therapeutic strategies.Several carcinogens have been identified in tobacco, and it is now well

stablished that there are differences in the individual ability to metabo-ize these products.73 It is conceivable that individuals exposed toersistently elevated levels of carcinogens, not just tobacco products, forprolonged period of time are at greater risk for tumorigenesis. Phase I

n the metabolism of tobacco components involves the activation ofrecursors through the CYP, cytochrome P450, enzyme systems, whereashase II involves detoxification of these products through conjugationechanisms. Although CYP-related polymorphisms exist, they have not

een found to significantly contribute to differences in risks for

68 Curr Probl Cancer, September/October 2004

Page 5: Prevention of head and neck cancer: Current status and future prospects

Si

ctaiIudit

C

isiaacuiBsbtagcdmg

dcnpdmd

C

CCHN.74-84 It is the phase II mechanisms which likely play a moremportant role in accounting for differential risks, as described below.The glutathione S-transferase (GST) genes are responsible for the

onjugation steps that abrogate tobacco toxin injury to DNA. Althoughhere are several components of the GST complex, GSTM1 and GSTT1re two well studied isoenzymes. It is now well recognized that there existndividual, racial, and ethnic differences in expressivity of this complex.ndividuals possessing the null genotype, in which the enzyme isnderexpressed or not expressed at all, are at a higher risk for theevelopment of squamous cell malignancies. This risk is higher than thatn controls when either of the isoenzymes is underexpressed and almostwice as high when both are underexpressed.85-95

hemopreventionA term first coined by Sporn and coworkers in 1976, chemoprevention

mplies the use of a natural or synthetic chemical for the reversal,uppression, or prevention of conversion of a premalignant lesion to annvasive form.96 This concept is of particular relevance to the uppererodigestive tract, where synchronous as well as metachronous lesionsre a significant cause of morbidity and mortality. The term “fieldancerization” was introduced by Slaughter and coworkers, who attrib-ted this to a field defect in the aerodigestive tract, which allowed forndependent generation of neoplastic clones at a number of sites.97

ladder cancer is the prototype of such skip lesions, but in SCCHN,imilar skip lesions have been identified. Interestingly, lesions surroundedy normal intervening mucosa share identical genetic abnormalities withhe primary or dominant invasive lesion. It is therefore hypothesized thatsingle cell first develops a clonal abnormality after sustaining a criticalenetic insult. It then migrates through the mucosa and deposits nests ofells in the adjacent regions, which tend to grow independently atifferent rates. Over a period of time, the dominant lesion becomesanifest as a malignancy, while others, although they share the same

enetic abnormalities, are not yet fully transformed.In accordance with this hypothesis, observations have been made byifferent investigators which add credence to the concept of fieldancerization. Califano and coworkers evaluated 18 patients with cervicalodal metastases (of squamous histology) from unknown head and neckrimary sites.98 In 10 of these patients, the investigators were able toemonstrate genetic abnormalities identical to the cervical lymph nodaletastases in benign appearing mucosa, and 3 of these patients eventually

eveloped invasive cancers.

urr Probl Cancer, September/October 2004 269

Page 6: Prevention of head and neck cancer: Current status and future prospects

CL

acaomol

hteShfiag

dtsnsa

Frap

2

linical Trials of Chemoprevention in Oral PremalignantesionsOral premalignant lesions have provided for an excellent model for

ssessing the efficacy of preventive agents. Several trials have beenompleted in patients with leukoplakia and erythroplakia. Leukoplakia isssociated with a less than 5% risk for malignant transformation, aspposed to erythroplakia, which carries a higher incidence, 30-50%, ofalignant transformation. The former has an approximately 30-50% rate

f spontaneous regression, in contrast to erythroplakia and dysplasticesions, where this rate is only 5%.104

The premise of chemopreventive efforts in the upper aerodigestive tractas been targeting of critical molecular lesions based on the aforemen-ioned observations. Although a number of genetic lesions, as outlinedarlier, have been identified in populations at risk for the development ofCCHN, and several others are yet to be discovered, most of the effortsave been targeted on the use of retinoids. Incidental observations wererst made on cattle deprived of vitamin A by Wolbach and Howe99; thesenimals had a much higher incidence of lung cancer and upper aerodi-estive tract malignancies.Retinoids have diverse biological effects, including the modulation ofifferentation, proliferation, and apoptosis in normal as well as malignantissues (Fig 1).100,101 Both the retinoid ligand and the receptor displayignificant diversity.102,103 There are at least two broad categories ofuclear retinoid receptors, RARs and RXRs, and each of them haveeveral subclasses. RXR ligands are more versatile since they can bind to

IG 1. Molecular model of retinoid chemoprevention. ATRA, all-trans-retinoic acid; 13cRA, 13-cisetinoic acid; RAR-beta, retinoic acid receptor beta. Adapted with permission from Cancer, Principlesnd Practice of Oncology, DeVita, Hellman, Rosenberg, Lippincott, Wilkins and Williams, 6th edition,587, 2001.

host of receptors, whereas RARs can only form heterodimers with

70 Curr Probl Cancer, September/October 2004

Page 7: Prevention of head and neck cancer: Current status and future prospects

Rrn

rorrhrsmdanli

Fapc

C

XRs. Some of the retinoids tested in clinical trials include 13-cis-etinoic acid (13cRA), 9-cis-retinoic acid, 4-N-4hydroxyphenyl reti-amide (fenretinide), bexarotene, and etretinate (Fig 2).Vitamin A Analogs. One of the pivotal studies testing the efficacy of

etinoids was completed in 1986 by Hong and coworkers.105 Patients withral premalignant lesions were randomized to high doses of 13-cis-etinoic acid at 1 to 2 mg/kg/day versus placebo. There was a 67%esponse rate with the use of the retinoid. However, toxicities (cheilitis,ypertriglyceridemia, erythema) were prohibitive during therapy, andelapses occurred frequently following cessation of therapy. This led to aeparate maintenance trial, in which patients were randomized, after 3onths of high-dose 13-cis-retinoic acid at 1.5 mg/kg/day, to either low

oses of maintenance 13-cis-retinoic acid at 0.5 mg/kg/day or �-carotenet 30 mg/day.106 This trial showed a significant advantage for mainte-ance with 13-cis-retinoic acid in comparison to �-carotene. However,ong-term follow up has failed to show an advantage for either

IG 2. Structures of selected retinoids and the carotenoid, beta-carotene. Retinol, 13-cis retinoic acid,nd beta-carotene are naturally occurring; 4HPR and etretinate are synthetic retinoids. Adapted withermission from Cancer, Principles and Practice of Oncology, DeVita, Hellman, Rosenberg, Lippin-ott, Wilkins and Williams, 6th edition, p576, 2001.

ntervention.

urr Probl Cancer, September/October 2004 271

Page 8: Prevention of head and neck cancer: Current status and future prospects

pstw1lsPI

wTvapIopsrodt3scaor

lrw3

a(dmc

2

Papadimitrakopolou and coworkers107 conducted a trial in which 36atients with advanced premalignant lesions (defined as moderate toevere dysplasia or carcinoma in situ) of the larynx and oral cavity werereated with a combination of �-interferon (3 � 106 IU/m2 three times aeek sq) with 13-cis-retinoic acid at 50 mg/m2/day and �-tocopherol at200 IU daily. Interestingly, almost 50% of the patients with laryngealesions demonstrated a complete reversal of their premalignant lesion, andeveral patients had long-lasting durable remissions for several months.atients with oral cavity lesions, however, did not have durable responses.n fact, disease progression occurred commonly.Stich and coworkers108 evaluated the effects of vitamin A in 65 patientsith oral leukoplakia in betel nut chewers and tobacco users in India.hirty-one of these patients were randomized to receive 100,000 IU ofitamin A and the other 34 placebo. Although 12 patients in the treatmentrm had responses, with no progressions reported, only 1 patient in thelacebo arm had a spontaneous regression, and the majority progressed.n a separate trial, the same investigators conducted a randomized studyf vitamin A administration at doses of 200,000 IU a week compared withlacebo in patients with leukoplakia. They reported a near 100%uppression in the incidence of leukoplakia, with almost a 50% completeesponse rate. Results of this study, as well as others, led to the conductf EUROSCAN, a large randomized study in patients with a prioriagnosis of head and neck cancer or lung cancer (see below). The first ofhree arms was randomized to the administration of vitamin A in doses of00,000 IU daily for the first year, followed by half the doses in theecond year. A second arm consisted of the above intervention inombination with N-acetylcysteine (600 mg) daily for 2 years, and a thirdrm of no intervention. The primary endpoint was assessment of the ratef SPT development. However, this trial failed to a show differences inecurrence rates, SPT development, or overall death rate.109

Chiesa and coworkers110,111 randomized 61 patients with oral prema-ignant lesions to either 4 months of etretinate (4-N-4hydroxyphenyletinamide/fenretinide) at 40 mg/day versus placebo. Major responsesere seen in 27 of the 31 patients treated with etretinate versus 5 of the0 patients in the placebo arm. Toxicity was minimal.The observations from some of the studies mentioned above are

ppealing from the standpoint of being able to identify an aberrant targetretinoic acid receptor) in SCCHN and being able to then disrupt theownstream signaling mediated by ligands (retinoids). Although theechanisms of vitamin A deficiency-induced carcinogenesis are not

learly understood, it is possible that this family of agents promote growth

72 Curr Probl Cancer, September/October 2004

Page 9: Prevention of head and neck cancer: Current status and future prospects

asmos

WcvtsbsEhpnpdr

mwSit

CC

tstr

refcba

C

nd differentiation through restoration of retinoic acid receptor-� expres-ion.112 Although a significant advance in our understanding of theechanisms of tumorigenesis in SCCHN, there are several limitations to

ur current approaches, which now warrant more complete, well-de-igned strategies in preventive approaches.Vitamin E. Vitamin E has been known to have antioxidant properties.hile most of the information pertaining to its effects in oral malignan-

ies is available from animal experiments, the mechanisms by whichitamin E may exert a protective function are speculated to be related tohis agent’s interaction with the immune system.113 Animal experimentsuggest a possible immunomodulatory role for �-tocopherol against bothacterial and viral infections.114 There are also epidemiological studies,imilar to those with selenium, which suggest a protective role for vitamin

as well, in geographic regions where the consumption of this agent isigher compared with other populations. These observations haverompted the investigation of vitamin E’s protective role against malig-ancies. There are limited data from human trials,115 in one of which 43atients with oral leukoplakia were treated with 400 IU of vitamin E twiceaily for 24 weeks. There were histological responses in 21% and clinicalegressions in the lesions in an additional 46% of the patients.116

Selenium. There are epidemiological observations that death rates fromalignancies, including SCCHN, are low in geographic regions of theorld where the soil is rich in selenium.117 The use of selenium inCCHN has been associated with an enhancement of cell-mediated

mmune responses. There are currently ongoing clinical trials evaluatinghe role of selenium in the adjuvant setting in lung cancer.118

linical Trials of Second Primary TumorhemopreventionFirst-Generation Trials. As mentioned previously, even after a curative

reatment, upper aerodigestive tract malignancies are associated with aignificant risk of recurrence; this is not limited to the local mucosa, buto the entire squamous epithelium which has been exposed to the sameisk factors.With this understanding, and based on their own observations of

eversibility of oral premalignant lesions, Hong and coworkers119 hypoth-sized that an intervention in upper aerodigestive tract malignanciesollowing adequate treatment (surgery/XRT) of SCCHN would be asso-iated with a reduction in the risk of second primary tumors. A doublelind, placebo-controlled trial was conducted of high-dose 13-cis-retinoic

cid (50-100 mg/m2/day) or placebo for 12 months. After a median

urr Probl Cancer, September/October 2004 273

Page 10: Prevention of head and neck cancer: Current status and future prospects

ftapleAti

cc(ep

orhms

wOSpsfINnnic

amscwr

2

ollow up of at least 32 months, there were significantly fewer SPTs in thereatment arm (4%) than in the placebo arm (24%; P � .005). In the samenalysis, the majority of the SPTs occurred in the epithelium at risk fromrior exposure to tobacco (93%), including the upper aerodigestive tract,ungs, and esophagus. However, similar to the observations made inarlier trials, toxicities (dermatological, gastrointestinal) were limiting.pproximately one third of the patients required discontinuation of

herapy due to these toxicities. Additionally, overall survival was notmpacted.After a median follow up of 4.5 years in this trial, Benner and

oworkers120 determined from analyzing these data that treated patientsontinued to have significantly fewer SPTs (14%) versus the placebo arm31%; P � .042). Most of this advantage actually occurred in thepithelium at risk, rather than at distant sites, and although this differenceersisted for as long as 2 years, it was no longer significant at 3 years.Bolla and coworkers,121 with a similar hypothesis, assessed the efficacyf etretinate in patients with a prior history of SCCHN. Patients wereandomized to either the retinoid at 50 mg/day for 1 month followed byalf the dose for 24 months or placebo. After a median follow up of 41onths, there was no difference in outcome, including the incidence of

econd primary tumors.Second-Generation Trials. Based on observations by Hong et al, asell as those by Pastorino and coworkers109 in lung cancer, the Europeanrganization for Research and Treatment of Cancer initiated the EURO-CAN trial. This was a large, multiinstitutional randomized study of 2592atients with completely resected/adequately treated stage I and II nonmall cell lung cancer as well as stages I, II, and III SCCHN. In a 2 � 2actorial design, patients were randomized on retinyl palmitate at 300,000U daily for 1 year followed by 150,000 IU in the second year, or to-acetylcysteine at 600 mg/d ayfor 2 years, or to both compounds, or too intervention at all. After a median follow up of 49 months, there wereo differences in outcome in any of the arms. The authors concluded thatnterventions consisting of 2 years of either retnyl palmitate or N-acetyl-ysteine did not provide protection from SPTs.In 1991, the University of Texas MD Anderson Cancer Center launchedrandomized, double blind trial of low-dose 13-cis-retinoic acid (30g/day) versus placebo to prevent SPTs in patients adequately treated for

tages I and II SCCHN. Of the 1384 patients randomized, 1189 wereonsidered eligible and evaluable. This study indicated that although thereas a transient protective effect of low-dose 13cRA on prevention of

ecurrence while the patients were on the study, there was no impact on

74 Curr Probl Cancer, September/October 2004

Page 11: Prevention of head and neck cancer: Current status and future prospects

ttio

aowAyrptnain

F

pcemrc

unawtmd

p

olm

C

he primary endpoint of second primary tumor prevention between thereatment and the placebo arm. Interestingly, however, continued smok-ng correlated with a poor outcome in these patients in terms of both ratef SPT development and survival.122,123

In a phase II clinical trial of biochemoprevention, Shin and coworkersdministered isoretinoin (50 mg/m2 orally daily), �-tocopherol (1200 IUrally daily), and IFN-� (3 � 106 units subcutaneously three times aeek) for 12 months in patients with treated locally advanced SCCHN.fter a median follow up of 49.4 months, the DFS rates at 1, 3, and 5ears were reported at 89, 78, and 74%, respectively. The overall survivalates at 1, 3, and 5 years were 98, 89, and 81%, respectively. Eighty-sixercent of the patients completed their therapy, suggesting acceptableolerability and compliance. Toxicities included weakness, peripheraleuropathy, as well as one report of optic neuritis. Based on these results,phase III randomized trial has been designed with reduced doses of

soretinoin (40 mg/m2 orally daily) and IFN-� (2 � 106 units subcuta-eously three times a week).124

uture DirectionsThere is now irrefutable evidence that one of the most effectivereventive interventions, particularly for SCCHN, is cigarette smokingessation. It is imperative that this important aspect be continuallymphasized by health care professionals to their patients. There isounting evidence that even in patients who have developed tobacco-

elated malignancies, smoking cessation results in an improved outcomeompared with those subjects who continue to smoke.For patients at risk for the development of SCCHN, as well as otherpper aerodigestive tract malignancies, a large-scale, concerted effort isecessary to better elucidate the mechanisms underlying carcinogenesis,s well as the development of targeted/molecular interventions whichould permit reversal or abrogation of the carcinogenic process. Al-

hough extensive work has been done to address some of these issues, asentioned previously, this effort should be used as a foundation for the

esign and execution of well-designed international trials.The following is an outline of potential candidate agents, as well asrospective studies.H-ras, a member of the ras gene family, is commonly altered in a varietyf solid tumors, including SCCHN. Approximately 30% of leukoplakiaesions and almost 50% of invasive SCCHN are associated with H-ras

utations. The role of farnesyl transferase inhibitors, which inhibit the

urr Probl Cancer, September/October 2004 275

Page 12: Prevention of head and neck cancer: Current status and future prospects

eI

tncvaieceNtios

Fe

2

nzymatic activation of H-ras, is currently being evaluated in phase I andI clinical trials, in both SCCHN and lung cancer.125-127

Cyclo-oxygenase, an enzyme that catalyzes the the synthesis of pros-aglandins, has been found to be overexpressed in a variety of premalig-ant and malignant states,128 including leukoplakia and SCCHN. In-reased levels of COX-2 may contribute to carcinogenesis through aariety of mechanisms, including the modulation of apoptosis andngiogenesis (Figs 3 and 4). In experimental models, COX-2 inhibitorsnhibit the carcinogenic process and suppress metastases, as well asnhance the effects of radiation therapy and chemotherapy. Lee andoworkers determined that at least seven SCCHN cell lines constitutivelyxpressed COX-2. They then cultured these cell lines in the presence ofS-398 and indomethacin, both of which are known to be inhibitors of

he COX-2 enzyme. After 72 h, there was a significant dose-dependentnhibition of cell growth, as well as a significant increase in the numberf cells that transitioned into the G0/G1 phase. Likewise, there was a

IG 3. COX 1 and 2 mediated prostaglandin synthesis. Adapted with permission from Dannenbergt al., 2002.

ignificant decrease in the number of cells in the S phase compared with

76 Curr Probl Cancer, September/October 2004

Page 13: Prevention of head and neck cancer: Current status and future prospects

ti

vAi(

oofELwPomyscrw

ihaccr

FIsP

C

hat in controls. Additionally, NS-398 was capable of inducing apoptosisn two of the three cell lines.Wang and coworkers demonstrated a significant reduction of newasculature development at tumor sites in mice treated with celecoxib.129

dditionally, there are reports of possible synergism between COX-2nhibitors and agents that target the epidermal growth factor receptorEGFR)130 (see below).The EGFR is frequently overexpressed in SCCHN. 131 Additionally,verexpression of EGFR is associated with a poor outcome, including thebservation of more aggressive growth in such tumors.132 The EGFRamily consists of at least four closely related growth factors, includingGFR/Her-1/erb-B1, Her-2/erb-B2/neu, Her-3/erb-B3, and Her-4/erb-B4.igand binding to EGFR results in the initiation of cytoplasmic signaling,hich is mediated through several different effectors, including Ras andI3K (phosphatidylinositiol-3-kinase). Downstream of this activation,ne of the signaling events involves phosphorylation of Akt, which in turnediates its antiapoptotic effect through inactivation (through phosphor-

lation) of BAD. Experiments with agents targeted to the EGFR havehown an abrogation of Akt phosphorylation and therefore increasedytotoxicity. Gupta and coworkers133 observed a significantly enhancedadiation-induced cytotoxic effect in patients with SCCHN when theyere also treated with gefitinib, an agent directed to the EGFR.A combined approach with the use of EGFR inhibitors and a COX-2

nhibitor has been explored by Shin and coworkers.134 The authorsypothesized that this combination may provide synergistic activitygainst SCCHN. In separate experiments, they treated two head and neckancer cell lines with gefitinib, an EGFR tyrosine kinase inhibitor alone,elecoxib alone, or the combination. Synergism with the combination was

134

IG 4. Levels of COX-2 protein are increased in HNSCC, as shown in this experiment, wheremmunoblotting was performed on HNSCC from six patients (odd lanes) and normal oral mucosa fromix healthy volunteers (even lanes). The immunoblot was probed with antibody specific for COX-2.urified ovine COX-2 was used as a standard. Reprinted with permission from Cancer Research.

eported. Based on these encouraging in vitro observations, the

urr Probl Cancer, September/October 2004 277

Page 14: Prevention of head and neck cancer: Current status and future prospects

imgcww

awaatgAtpo3

rgtppw

phafidtr

SwauiaH

2

nvestigators conducted similar experiments in nude mice (xenograftodels) in whom the Tu212 SCCHN cell line was introduced. Four

roups of animals were subjected to gefitinib alone, celecoxib alone, theombination, or a placebo vehicle. Significant synergism was reportedith the combination, leading the investigators to propose phase I/II trialsith this combination.135

Magne and coworkers136 tested the antiproliferative activity of gefitinibgainst SCCHN cell lines and were able to establish increasing activityith higher growth factor receptor levels. They furthered this research to

ssess the specific molecular mechanisms underlying this activity, as wells explored the possibility of synergism between gefitinib and chemo-herapeautic agents. They cultured CAL 33 cells, a SCCHN cell line, withefitinib, followed 48 h later with the addition of 5-FU and cisplatin.lthough gefitinib alone was able to induce accumulation of these cells in

he G0/G1 phase of the cell cycle, with a concomitant increase in p21,27, and BAX as well as a decrease in Akt phosphorylation, the additionf the chemotherapeutic agents resulted in a dramatic increase in caspaseactivity as well.137

Cohen and coworkers recently reported on 47 patients treated forecurrent SCCHN. About 11% of the patients responded to 500 mg ofeftinib administered daily.138 Although dermatological and gastrointes-inal toxicities are prominent, the reports of rare cases of interstitialneumonitis as well may limit the application of this strategy in areventive setting. There are ongoing trials of lower doses of geftinib,hich are not yet mature enough to report on toxicities or efficacy.Alterations in p53 are commonly reported in SCCHN. Due to the poorrognosis associated with p53 mutations, an ongoing trial, ONYX-015,as preliminarily reported on the use of p53 administered with andenoviral vector in patients with oral premalignant lesions. Two of therst 10 patients treated with weekly administration as a mouthwash haveemonstrated resolution of dysplasia.139 An ongoing study is evaluatinghe role of tumoral injections of the adenoviral vector in patients withecurrent SCCHN.With advances in our current understanding of the molecular biology ofCCHN, there are now several more experimental approaches availableith novel agents that may provide for more effective chemopreventive

pproaches. Future trials in chemoprevention will require a betternderstanding of the intermediate biomarkers of response, so thatnterventions can be continued, or interrupted, based on the prognosticnd predictive value of these intermediary biomarkers of response.

owever, it is imperative that the scientific community not repeat some

78 Curr Probl Cancer, September/October 2004

Page 15: Prevention of head and neck cancer: Current status and future prospects

oaaam

C

f the previous efforts that have resulted in suboptimal results. Singlegent interventions have not necessarily resulted in meaningful outcomes,nd the challenge will be to identify effective combinations of biologicalgents that would be safe, as well as effective in chemoprevention ofalignancies

REFERENCES1. Parkin DM, Pisani P, Ferlay J. Estimates of the worldwide incidence of 25 major

cancers in 1990. Objective evidence of SCCHN as a major cancer in South CentralAsia. Int J Cancer 1999;80:827-84.

2. Landis SH, Murray T, Bolden S, Wingo PA. Cancer Statistics, 1999. CA CancerJ Clin 1999;49:8-31.

3. Vokes EE, Stenson KM. Therapeutic options for laryngeal cancer. N Engl J Med2003;349:2087-9.

4. Vikram B. Changing patterns of failure in advanced head and neck cancer. ArchOtolryngol 1984;110:564-5.

5. Cooper JS, Pajak TK, Rubin P, et al. Second malignancies in patients who havehead and neck cancer: incidence, effect on survival and implications based onRTOG experience. Int J Radiat Oncol Biol Phys 1989;17:449-56.

6. Licciardello JT, Spitz MR, Hong WK. Multiple primary cancers in patients withcancer of the head and neck; second cancer of the head and neck, esophagus andlung. Int J Radiat Oncol Biol Phys 1989;17:467-76.

7. Gao X, Fisher SG, Mohideen N, et al. Second primary cancers in patients withlaryngeal cancer: a population based study (abstract). Proc Am Soc Clin Oncol2000;19:414a.

8. Tucker MA, Murray N, Shaw EG, et al. Second primary cancers related to smokingand treatment of small-cell lung cancer. J Natl Cancer Inst 1997;89:1782-6.

9. Russo A, Crosigani P, Berrino F. Tobacco smoking, alcohol drinking and dietaryfactors as determinants of new primaries among male laryngeal cancer patients: acase cohort study. Tumori 1996;82:510-25.

10. Kurokawa R, Direnzo J, Boehm M, et al. Regulation of retinoid signaling byreceptor polarity and allosteric control of ligand binding. Nature 1994;371:528.

11. Van Leeuwen FE, Klokman WJ, Stovall M, et al. Role of radiotherapy and smokingin lung cancer following Hodgkin’s Disease. J Natl Cancer Inst 1995;87:1530-7.

12. Richardson GE, Tucker MA, Venzon DJ, et al. Smoking cessation after successfultreatment of small-cell lung cancer is associated with fewer smoking-related secondprimary cancers. Ann Intern Med 1993;119:383-90.

13. Silverman S Jr, Grosky M, Greenspan D. Tobacco usage in patients with head andneck carcinomas: a follow up study on habit changes and second primaryoral/oropharyngeal cancers. J Am Dent Assoc 1983;106:33-5.

14. Castigliano SG. Influence of continued smoking on the incidence of second primarycancers involving mouth, pharynx and larynx. J Am Dent Assoc 1968;77:580-5.

15. Bishop JM. Molecular themes in oncogenes. Cell 1991;64:235.16. Berenson JR, Yan J, Micke RA. Frequent amplification of the bcl-1 locus in head

and neck squamous cell carcinomas. Oncogene 1989;41:111.

urr Probl Cancer, September/October 2004 279

Page 16: Prevention of head and neck cancer: Current status and future prospects

2

17. Callander T, El-Nagger AK, Lee MS, et al. PRAD-1 (CCND1)/cyclin D1 oncogeneamplification in primary head and neck squamous cell carcinoma. Cancer 1994;74:152.

18. Jares P, Fernandez PL, Campo E, et al. PRAD-1/cyclin D1 gene amplificationcorrelates with messenger RNA over-expression and tumor progression in humanlaryngeal carcinoma. Cancer Res 1994;54:4813.

19. Trink B, Okami K, Wu L, et al. A new p53 homologue. Nat Med 1998;4:74.20. Hibi K, Trink B, Patturajan M et al. AIS is an oncogene amplified in squamous cell

carcinoma. Proc Natl Assoc Sci (in press).21. Grandis JR, Tweardy DJ. Elevated levels of transforming growth factor alpha and

epidermal growth factor receptor messenger RNA are early markers of carcino-genesis in head and neck cancer. Cancer Res 1993;53:3579.

22. He Y, Zeng Q, Drenning SD, et al. Inhibition of human squamous cell carcinomagrowth in vivo by epidermal growth factor receptor anti-sense RNA transcribedfrom the U6 promoter. J Natl Cancer Inst 1998;90:1080.

23. Grandis JR, Drenning SD, Chakrobarty A, et al. Requirement of stat3 but not stat1activation for epidermal growth factor receptor-mediated cell growth in vitro. J ClinInvest 1998;102:1385.

24. Sorrells DL, Ghali GE, Meschonat C, et al. Competitive PCR to detect eIF4E geneamplification in head and neck cancer. Head Neck 1999;21:60.

25. Sorrels DL, Meschonat C, Black D, Li BD. Pattern of amplification and overex-pression of the eukaryotic initiation factor 4E gene in solid tumors. J Surg Res1999;85:37.

26. Nathan CA, Franklin S, Abreo FW, et al. Expression of eIF4E head and necktumorigenesis; possible role in angiogenesis. Laryngoscope 1999;109:1253.

27. Chan G, Boyle JO, Yang EK, et al. Cyclo-oxygenase-2 expression is up-regulatedin squamous cell carcinoma of the head and neck. Cancer Res 1999;59:991.

28. Lin DT, Subbaramaiah K, Shah JP, Dannenberg AJ, Boyle JO. Cyclooxygenase-2:a novel molecular target for the prevention and treatment of head and neck cancer.Head Neck 2002;24:792-9.

29. Kujubu DA, Fletcher BS, Varnum BC, et al. TIS10, a phorbol ester tumorpromoter-inducible mRNA from Swiss 3T3 cells, encodes a novel prostaglandinsynthase/cyclooxygenase homologue. J Biol Chem 1991;266:12866-72.

30. DuBois RN, Awad J, Morrow J, et al. Regulation of eicosanoid production andmitogenesis in rat intestinal epithelial cells by transforming growth factor- andphorbol ester. J Clin Invest 1994;93:493-8.

31. Jones DA, Carlton DP, McIntyre TM, et al. Molecular cloning of humanprostaglandin endoperoxide synthase type II and demonstration of expression inresponse to cytokines. J Biol Chem 1993;68:9049-54.

32. Rimarachin JA, Jacobson JA, Szabo P, et al. Regulation of cyclooxygenase-2expression in aortic smooth muscle cells. Arterioscler Thromb 1994;14:1021-31.

33. Kelley DJ, Mestre JR, Subbaramaiah K, et al. Benzo[a]pyrene upregulatescyclooxygenase-2 gene expression in oral epithelial cells. Carcinogenesis 1997;18:795-9.

34. Xie W, Herschman HR. v-src induces prostaglandin synthase 2 gene expression byactivation of the c-Jun N-terminal kinase and the c-Jun transcription factor. J Biol

Chem 1995;270:27622-8.

80 Curr Probl Cancer, September/October 2004

Page 17: Prevention of head and neck cancer: Current status and future prospects

C

35. Subbaramaiah K, Telang N, Ramonetti JT, et al. Transcription of cyclooxygenase-2is enhanced in transformed mammary epithelial cells. Cancer Res 1996;56:4424-9.

36. Subbaramaiah K, Altorki N, Chung WJ, et al. Inhibition of cyclooxygenase-2 geneexpression by p53. J Biol Chem 1999;274:10911-15.

37. Boyle JO, Hakim J, Koch W, et al. The incidence of p53 mutations increases withprogression of head and neck cancer. Cancer Res 1993;53:4477-80.

38. Eberhart CE, Coffey RJ, Radhika A, et al. Up-regulation of cyclooxygenase 2 geneexpression in human colorectal adenomas and adenocarcinomas. Gastroenterology1994;107:1183-8.

39. Wilson KT, Fu S, Ramanujam KS, Meltzer SJ. Increased expression of induciblenitric oxide synthase and cyclooxygenase-2 in Barrett’s esophagus and associatedadenocarcinomas. Cancer Res 1998;58:2929-34.

40. Zimmermann KC, Sarbia M, Weber AA, et al. Cyclooxygenase-2 expression inhuman esophageal carcinoma. Cancer Res 1999;59:198-204.

41. Shiota G, Okubo M, Noumi T, et al. Cyclooxygenase-2 expression in hepatocellularcarcinoma. Hepatogastroenterology 1999;46:407-12.

42. Ristimaki A, Honkanen N, Jankala H, et al. Expression of cyclooxygenase-2 inhuman gastric carcinoma. Cancer Res 1997;57:1276-80.

43. Soslow RA, Dannenberg A, Baergen RN, et al. Cyclooxygenase-2 expression isupregulated in common colonic, pulmonary and mammary tumors. Cancer 2000;89:2637-48.

44. Chan G, Boyle JO, Yang EK, et al. Cyclooxygenase-2 expression is up-regulatedin squamous cell carcinoma of the head and neck. Cancer Res 1999;59:991-4.

45. Gupta S, Srivastava M, Ahmad N, et al. Over-expression of cyclooxygenase-2 inhuman prostate adenocarcinoma. Prostate 2000;42:73-8 Links.

46. Wolff H, Saukkonen K, Anttila S, et al. Expression of cyclooxygenase-2 in humanlung carcinoma. Cancer Res 1998;58:4997-5001.

47. Tucker ON, Dannenberg AJ, Yang EK, et al. Cyclooxygenase-2 expression isup-regulated in human pancreatic cancer. Cancer Res 1999;59:987-90 Links.

48. Mohammed SI, Knapp DW, Bostwick DG, et al. Expression of cyclooxygenase-2(COX-2) is human invasive transitional cell carcinoma (TCC) of the urinarybladder. Cancer Res 1999;59:5647-50.

49. Kulkarni S, Rader JS, Zhang F, et al. Cyclooxygenase-2 is overexpressed in humancervical cancer. Clin Cancer Res 2001;7:429-34.

50. Gallo O, Franchi A, Magnelli L, et al. Cyclooxygenase-2 pathway correlates withVEGF expression in head and neck cancer. Implications for tumor angiogenesis andmetastasis. Neoplasia 2001;3:53-61.

51. Wang X, Pavelic ZP, Li Y, et al. Overexpression and amplification of glutathioneS-transferase pi gene in head and neck squamous cell carcinomas. Clin Cancer Res1997;3:111.

52. Nawroz H, van der Riet P, Hruban RH, et al. Allelotype of head and neck squamouscell carcinoma. Cancer Res 1994;54:1152.

53. Ahsee KW, Cooke TG, Pickford IR, et al. An allelotype of squamous carcinoma ofthe head and neck using microsatellite markers. Cancer Res 1994;54:1617.

54. van der Riet P, Nawroz H, Hruban RH, et al. Frequent loss of chromosome 9p21

early in head and neck cancer progression. Cancer Res 1994;54:1156.

urr Probl Cancer, September/October 2004 281

Page 18: Prevention of head and neck cancer: Current status and future prospects

2

55. Zhang SY, Kleinszanto AJP, Sauter ER, et al. Higher frequency of alterations in thep16/CDKN2 gene in squamous cell carcinoma cell lines than in primary tumors ofthe head and neck. Cancer Res 1994;54:5050.

56. Cairns P, Mao L, Merlo A, et al. Low rate of p16 (MTS1) mutations in primarytumors with 9p loss. Science 1994;265:415.

57. Zhang Y, Xiong Y, Yarbrough WG. ARF promotes MDM2 degradation andstabilizes p53: ARF-INK4a locus deletion impairs both the Rb and p53 tumorsuppression pathways. Cell 1998;92:725.

58. Pomerantz J, Schreiber-Agus N, Liegeois NJ, et al. The INK4a tumor suppressorgene product, p19**ARF, interacts with MDM2 and neutralizes MDM2’s inhibi-tion of p53. Cell 1998;92:713.

59. Liggett WH Jr, Sewell DA, Rocco J, et al. p16 and p16beta are potent growthsuppressors of head and neck squamous carcinoma cells in vitro. Cancer Res1996;56:4119.

60. Izzo JG, Papadimitrakopoulou VA, Liu DD, et al. Cyclin D1 genotype, response tobiochemoprevention, and progression rate to upper aerodigestive tract cancer.J Natl Cancer Inst 2003;95:198-205.

61. Maestro R, Gasparotto D, Vuksavljevic T, et al. Three discrete regions of deletionin head and neck cancers. Cancer Res 1993;53:5775.

62. Shin DM, Kim J, Ro JY, Hittelman J, et al. Activation of p53 gene expression inpremalignant lesions during head and neck tumorigenesis. Cancer Res 1994;54:321-6.

63. Lippman SM, Shin DM, Lee JJ, et al. p53 and retinoid chemoprevention of oralcarcinogenesis. Cancer Res 1995;55:16-9.

64. Shin DM, Lee JS, Lippman SM, et al. p53 expressions: predicting recurrence andsecond primary tumors in head and neck squamous cell carcinoma. J Natl CancerInst 1996;88:519-29.

65. Wu CL, Sloan P, Read AP, et al. Deletion mapping on the short arm ofchromosome 3 in squamous cell carcinoma of the oral cavity. Cancer Res1994;54:6484.

66. Okami K, Wu L, Riggins G, et al. Analysis of PTEN/MMAC1 alterations inaerodigestive tract tumors. Cancer Res 1998;58:509.

67. Shao X, Tandon R, Samara G, et al. Mutational analysis of the PTEN gene in headand neck squamous cell carcinoma. Int J Cancer 1998;77:684.

68. Yoo GH, Xu HJ, Brennan JA, et al. Infrequent inactivation of the retinoblastomagene despite frequent loss of chromosome 13q in head and neck squamous cellcarcinoma. Cancer Res 1994;54:4603.

69. Goldgar DG, Easton DF, Cannon-Albright LA, et al. Systematic population basedassessment of cancer risk in first degree relatives of cancer probands. J Natl CancerInst 1994;86:1600-8.

70. Foulkes WD, Brunet JS, Kowalski LP, et al. Family history of cancer is a risk factorfor squamous cell cancer of the head and neck in Brazil: a case controlled study. IntJ Cancer 1995;63:769-73.

71. Copper MP, Jovanovic A, Nauta JJP, et al. Role of genetic factors in the etiologyof squamous cell carcinoma of the head and neck. Arch Otolaryngol Head NeckSurg 1995;121:157-60.

72. Foulkes WD, Brunet JS, Sieh W, et al. Familial risks of squamous cell carcinoma

of the head and neck: retrospective case control study. Br Med J 1996;313:716-21.

82 Curr Probl Cancer, September/October 2004

Page 19: Prevention of head and neck cancer: Current status and future prospects

C

73. Wei Q, Xu X, Cheng L, et al. Simultaneous amplification of four DNA repair genesand beta-actin in human lymphocytes by multiplex reverse transcriptase-PCR.Cancer Res 1995;55:5025-9.

74. Wei Q, Eicher SA, Guan Y, et al. Reduced expression of hMLH1 and hGTBP/hMSH6: a risk factor for head and neck cancer. Cancer Epidemiol Biomarkers Prev1998;7:309-14.

75. Cheng L, Sturgis EM, Eicher SA et al. Nucleotide excision repair gene expressionand the risk for SCCHN. Cancer (in press).

76. Cheng L, Eicher SA, Guo Z, et al. Reduced DNA repair capacity in head and neckcancer patients. Pilot study demonstrating decreased repair ability in SCCHNpatients as compared to cancer-free control subjects. Cancer Epidemiol BiomarkersPrev 1998;7:465-8.

77. Papadimitrakopoulu V. Chemoprevention of head and neck cancer: an update. CurrOpin Oncol 2002;14:318-22.

78. Li D, Firozi PF, Chang P, et al. In vitro BPDE -induced DNA adducts in peripherallymphocytes as a risk factor for squamous cell carcinoma of the head and neck. IntJ Cancer 2001;93:436-40.

79. Pindborg JJ, Murti PR, Bhonsle RB, et al. Oral submucus fibrosis as a precancerouscondition. Scand J Dent Res 1984;92:224-9.

80. Murti PR, Bhonsle RB, Pindborg JJ, et al. Malignant transformation rate in oralsubmucuc fibrosis over a 17 year period. Community Dent Oral Epidemiol1985;13:340-1.

81. Murti PR, Bhonsle RB, Gupta PC, et al. Etiology of submucus oral fibrosis withspecial reference to the role of areca nut chewing. J Oral Pathol Med 1885;24:145-52.

82. Shen H, Sturgis EM, Khan SG, et al. An intronic poly (AT) polymorphism of theDNA repair gene XPC and risk of squamous cell carcinoma of the head and neck:a case controlled study. Cancer Res 2001;61:3321-5.

83. Shen MR, Jones IM, Mohrenweiser H. Nonconservative amino acid substitutionvariants exist at polymorphic frequency in DNA repair genes in healthy humans.Cancer Res 1998;58:604-8 Initial study screening cancer-free subjects for SNPs ofDNA repair genes.

84. Sturgis EM, Castillo EJ, Li L, et al. Polymorphisms of the DNA repair geneXRCC1 in squamous cell carcionoma of the head and neck. Carcinogenesis1999;20:2125-9 Initial study demonstrating the feasibility of DNA repair geneSNPs as markers of SCCHN susceptibility.

85. Sturgis, ErichM, WeiQingyi. Genetic susceptibility—molecular biology of headand neck cancer. Curr Opin Oncol 2002;14:310-17.

86. Sturgis EM, Castillo EJ, Li L, et al. Polymorphisms of XPD/ERCC2 exon 8 areextremely rare and not a significant risk factor for squamous cell carcinoma of thehead and neck. Oral Oncol (in press).

87. Sturgis EM, Zheng R, Li L, et al. XPD/ERCC2 polymorphisms and risk of head andneck cancer: a case-control analysis. Carcinogenesis 2000;21:2219-23.

88. Sugasawa K, Ng JM, Masutani C, et al. Xeroderma pigmentosum group C proteincomplex is the initiator of global genome nucleotide excision repair. Mol Cell

1998;2:223-32.

urr Probl Cancer, September/October 2004 283

Page 20: Prevention of head and neck cancer: Current status and future prospects

1

1

1

1

1

1

1

1

2

89. Jahnke V, Matthias C, Fryer A, et al. Glutathione S-transferase and cytochromeP-450 polymorphism as risk factors for squamous cell carcinoma of the larynx.Am J Surg 1996;172:671-3.

90. Benhamou S, Bouchardy C, Paoletti C, et al. Effects of CYP2D6 activity andtobacco on larynx cancer risk. Cancer Epidemiol Biomarkers Prev 1996;5:683-6.

91. Lucas D, Menez C, Floch F, et al. Cytochromes P4502E1 and P4501A1 genotypesand susceptibility to cirrhosis or upper aerodigestive tract cancer in alcoholicCaucasians. Alcohol Clin Exp Res 1996;20:1033-7.

92. Park JK, Muscat JE, Ren Q, et al. CYP1A1 and GSTM1 polymorphisms and oralcancer risk. Cancer Epidemiol Biomarkers Prev 1997;6:791-7.

93. Morita S, Yano M, Tsujinaka T, et al. Genetic polymorphisms of drug-metabolizingenzymes and susceptibility to head-and-neck squamous-cell carcinoma. Int JCancer 1999;80:685-8.

94. Gonzales MV, Alvarez V, Pello MF, et al. Genetic polymorphism of N-acetyl-transferase-2, glutathione S-transferase-M1, and cytochromes P450IIE1 andP450IID6 in the susceptibility to head and neck cancer. Clin Pathol 1998;51:294-8.

95. Ophuis MBO, van Lieshout EMM, Roelofs HMJ, et al. Glutathione S-transferaseM1 malignant head and neck lesions. Cancer 1998;82:936-43.

96. Sporn MB, Dunlop NM, Newton DL, Smith JM. Prevention of chemical carcino-genesis by vitamin A and its synthetic analogs. Fed Proc 1976;35:1332-8.

97. Slaughter DP, Southwick HW, Smejkal W. Field cancerization in oral stratifiedsquamous epithelium: clinical implications of multicentric origin. Cancer 1953;6:963-8.

98. Califano J, Westra WH, Meninger G, et al. Unknown primary head and necksquamous cell carcinoma: molecular identification of the site of origin. J NatlCancer Inst 1999;91:599.

99. Wolbach SB, Howe PR. Tissue changes following deprivation of fat solublevitamin A. J Exp Med 1925;42:753.

00. Manglesdorf DJ, Umesono K, Evans RM. The retinoid receptors. In: Sporn MB,Roberts AB, Goodman DS, editors. The retinoids. New York: Raven Press,1994:319-49.

01. Chambon P. The retnoid signaling pathway: molecular and genetic analyses. SeminCell Biol 1994;5:115-25.

02. Leid M, Kastner P, Chambon P. Multiplicity generates diversity in the retinoic acidsignaling pathways. Trends Biochem Sci 1992;17:427-33.

03. Blaner WS. Biochemistry and pharmacology of retinoids. In: Hong WK, Lotan R,editors. Retinoids in oncology. New York: Dekker, 1993:1-42.

04. Stich HR, Rosin MP, Hornby P, et al. Remission of oral leukoplakias andmicronuclei in tobacco/betel quid chewers treated with beta-carotene plus vitaminA. Intl J Cancer 1988;42:195-9.

05. Hong W, Endicott J, Itri LM, et al. 13-cis-Retinoic acid in the treatment of oralleukoplakia. N Engl J Med 1986;315:1501-5.

06. Lippman SM, Batsakis JG, Toth BB, et al. Comparison of low dose isoretinoin withbeta carotene to prevent oral carcinogenesis. N Engl J Med 1993;328:15-20.

07. Papadimitrikopoulou VA, Hong WK, Lee JS, et al. Low dose isoretinoin versusbeta carotene to prevent oral carcinogenesis: long term follow up. J Natl Cancer Inst

1997;89:257-8.

84 Curr Probl Cancer, September/October 2004

Page 21: Prevention of head and neck cancer: Current status and future prospects

1

1

1

1

1

11

1

1

11

1

1

1

1

1

1

C

08. Stich HF, Hornby AP, Mathew B, et al. Response of oral leukoplakias to theadministration of vitamin A. Cancer Lett 1988;40:93-101.

09. van Zandwijk N, Dalesio O, Pastorino U, et al. EUROSCAN, a randomized trial ofvitamin A and N-acetylcysteine in patients with head and neck cancer or lungcancer. For the European Organization for Research and Treatment of Cancer of theHead and Neck and Lung Cancer Cooperative Groups. J Natl Cancer Inst2000;92:977-86.

10. Chiesa F, Tradati N, Marazza M, et al. Prevention of local relapses and newlocalizations of oral leukoplakias with the synthetic retinoid fenretinide (4-HPR).Preliminary results. Eur J Cancer B Oral Oncol 1992;28B:97-102.

11. Chiesa F, Tradati N, Marazza M, et al. Fenretinide (4HPR) in chemoprevention oforal leukoplakia. J Cell Biochem Suppl 1993;17F:255-61.

12. Lotan R, Xu XC, Lippman SM, et al. Suppression of retinoic acid receptor-beta inpremalignant oral lesions and its upregulation by isoretinoin. N Engl J Med1995;332:1405-10.

13. Moriguchi S, Muraga M. Vitamin E and immunity. Vitam Horm 2000;59:305-36.14. Prasad JS. Effect of vitamin E supplementation on leukocyte function. Am J Clin

Nutr 1980;33:606-8.15. Anonymous. The effect of vitamin E and beta carotene on the incidence of lung

cancer and other cancers in male smokers. The Alpha-Tocopherol, Beta CaroteneCancer Prevention Study Group. N Engl J Med 1994;330:1029-35.

16. Malvy DJ, Favier A, Faure H, et al. Effect of two years’ supplementation withnatural antioxidants on vitamin and trace element status biomarkers: preliminarydata of the SU.VI.MAX study. Cancer Detect Prev 2001;25:479-85.

17. Patterson BH, Levander OA. Cancer Epidemiol Biomarkers Prev 1997;6:63-9.18. Klein EA, Thompson IM, Lippman SM, et al. SELECT the next prostate cancer

prevention trial: Selenium and Vitamin E Cancer Prevention Trial. J Urol2001;166:1311-1315.

19. Hong WK, Lippman SM, Itri LM, et al. Prevention of second primary tumors withisoretinoin in squamous cell carcinoma of the head and neck. N Engl J Med1990;323:795-801.

20. Benner SE, Pajak TF, Lippman SM. Prevention of second primary tumors withisoretinoin in patients with squamous cell carcinoma of the head and neck: longterm follow up. J Natl Cancer Inst 1994;86:141.

21. Bolla M, Lefur R, Ton Van J, et al. Prevention of second primary tumors withetretinate in squamous cell carcinoma of the oral cavity and oropharynx. Results ofa multicentric double-blind randomized study. Eur J Cancer 1994;30A:767-72.

22. Khuri FR, Kim ES, Lee JJ. The impact of smoking status, disease stage and indextumor site on second primary tumor incidence and tumor recurrence in the head andneck retinoid chemoprevention trial. Cancer Epidemiol Biomarkers Prev 2001;10:823-9.

23. Khuri FR, Lee JJ, Lippman SM. Isoretinoin effects on head and neck cancerrecurrence and second primary tumors. Proceedings of the American Society ofOncology 2003;22:359a.

24. Shin DM, Khuri FR, Murphy B, et al. Combined interferon alpha, 13-cis-retinoicacid and alpha tocopherol in locally advanced head and neck squamous cell

carcinoma: novel bioadjuvant phase II trial. J Clin Oncol 2001;19:3010-17.

urr Probl Cancer, September/October 2004 285

Page 22: Prevention of head and neck cancer: Current status and future prospects

1

1

1

1

1

1

1

1

1

1

1

1

1

1

1

2

25. Anderson JA, Irish JC, McLachlin CM. H-ras oncogene mutation and humanpapillomavirus infection in oral carcinomas. Arch Otolaryngol Head Neck Surg1994;120:755-60.

26. Oku N, Shimada K, Itoh H. Ha-ras oncogene product in human oral squamous cellcarcinoma. Kobe J Med Sci 1989;35:277-86.

27. Hahn SM, Bernhard EJ, Regine W. A phase I trial of the farnesyl-transferaseinhibitorL-778,123 and radiotherapy for locally advanced lung and head and neckcancer. Clin Cancer Res 2002;8:1065-72.

28. Lin DT, Subaramaiah K, Shah JP. Cyclo-oxygenase-2: a novel molecular target forthe prevention and treatment of head and neck cancer. Head Neck 2002;24:792-9.

29. Wang Z, Fuentes CF, Shapshay SM. Antiangiogenic and chemopreventive activi-ties of celecoxib in oral carcinoma cell. Laryngoscope 2002;112:839-43.

30. Chen Z, Zhang X, Wang. A possible interaction between epidermal growth factorreceptor and cyclooxygenase-2 mediated pathways in squamous cell carcinoma ofthe head and neck. Proceedings of the American Association of Cancer Research2003;44:510a.

31. Shin DM, Ro JY, Hong WK, Hittleman WN. Dysregulation of epidermal growthfactor receptor expression in premalignant lesions during head and neck tumori-genesis. Cancer Res 1994;54:3153-9.

32. Grandis JR, Melhem MF, Gooding WE, et al. Levels of TGF alpha and EGFRprotein in head and neck squamous cell carcinoma and patient survival. J NatlCancer Inst 1998;90:824-32.

33. Anjali K, Gupta W, Gillies McKenna CN, et al. Local recurrence in head and neckcancer: relationship to radiation resistance and signal transduction. Clin Cancer Res2002;8:885-92.

34. Shin DM, Wang ZQ, Li M, et al. ZD 1839, an epidermal growth factor receptortyrosine kinase inhibitor (TKI), inhibits cell growth synergistically with a cylooxy-genase-2 (COX-2) inhibitor, celecoxib, in squamous cell carcinoma of the head andneck (SCCHN). Proceedings of the American Association of Cancer Research2003;44:1084:4729a.

35. Zhang X, Chen Z, Shin DM, et al. Significant delay of tumor progression by ZD1839, an epidermal growth factor receptor tyrosine kinase inhibitor (EGFR-TKI), incombination with a cyclooxygenase-2 (COX-2) inhibitor, Celecoxib, in a xenograftmodel for squamous cell carcinoma of the head and neck. Proceedings of theAmerican Association of Cancer Research 2004;45:726:3136a.

36. Magne N, Fischell JL, Dubreuil A, et al. Influence of epidermal growth factorreceptor (EGFR), p53 and intrinsic MAP Kinase pathway status of tumour cells onthe antiproliferative effect of ZD1839 (Iressa). Br J Cancer 2002;86:1518-23.

37. Magne N, Fischell JL, Tiffon C, et al. Molecular mechanisms underlying theinteraction between ZD 1839 (Iressa) and cisplatin and 5-FU. Br J Cancer2003;89:585-92.

38. Cohen E, Rosen F, Stadler WM, et al. Phase II trial of ZD1839 in recurrent ormetastatic squamous cell carcinoma of the head and neck. J Clin Oncol 2003;21:1980-7.

39. Cohen E, Papadimitrakopolou V, Silverman S, et al. A phase II trial of anattenuated adenovirus, ONYX-015, as mouthwash therapy for premalignant oraldysplasia. Proceedings of the American Society of Clinical Oncology 2001;20:229a.

86 Curr Probl Cancer, September/October 2004