molecular origin lung cancer

Upload: darmarianto

Post on 02-Jun-2018

217 views

Category:

Documents


0 download

TRANSCRIPT

  • 8/10/2019 Molecular Origin Lung Cancer

    1/14

    T h e n e w e n g l a n d j o u r n a l o f medicine

    n engl j med 359;13 www.nejm.org september 25, 2008 1367

    review article

    Molecular Origins of Cancer

    Lung Cancer

    Roy S. Herbst, M.D., Ph.D., John V. Heymach, M.D., Ph.D.,and Scott M. Lippman, M.D.

    From the Departments of Thoracic/Headand Neck Medical Oncology (R.S.H.,J.V.H., S.M.L.), Cancer Biology (R.S.H.,J.V.H.), and Clinical Cancer Prevention(S.M.L.), University of Texas M.D. Ander-son Cancer Center, Houston. Address re-print requests to Dr. Lippman at the De-partment of Thoracic/Head and NeckMedical Oncology, Unit 432, M.D. Ander-

    son Cancer Center, 1515 Holcombe Blvd.,Houston, TX 77030, or at [email protected].

    Drs. Herbst and Heymach contributedequally to this article.

    N Engl J Med 2008;359:1367-80.Copyright 2008 Massachusetts Medical Society.

    Lung cancer is the leading cause of cancer deaths in the united

    States1and worldwide. The two major forms of lung cancer are nonsmall-cell lung cancer (about 85% of all lung cancers) and small-cell lung cancer

    (about 15%). Despite advances in early detection and standard treatment, nonsmall-cell lung cancer is often diagnosed at an advanced stage and has a poorprognosis. The treatment and prevention of lung cancer are major unmet needs thatcan probably be improved by a better understanding of the molecular origins and

    evolution of the disease.Nonsmall-cell lung cancer can be divided into three major histologic subtypes:

    squamous-cell carcinoma, adenocarcinoma, and large-cell lung cancer. Smokingcauses all types of lung cancer but is most strongly linked with small-cell lungcancer and squamous-cell carcinoma; adenocarcinoma is the most common typein patients who have never smoked (Fig. 12-8). This review will focus on majorrecent advances in the molecular study of the origins and biology of squamous-cellcarcinoma and adenocarcinoma, since they constitute the vast majority of diagnosedlung cancers (Table 15,8-14). These advances have been facilitated by the developmentof molecular techniques and biomarkers for defining cancer risk, prognosis, andoptimal therapy aimed at personalized prevention and treatment of lung cancer.

    Molecular Origins

    Host Susceptibility

    Epidemiologic studies showing an association between family history and an in-creased risk of lung cancer provided the first evidence of host susceptibility (Fig. 1).Lung-cancer susceptibility and risk also are increased in inherited cancer syndromescaused by rare germ-line mutations in p53,15retinoblastoma,16and other genes,17as well as a germ-line mutation in the epidermal growth factor receptor (EGFR)gene.18More recently, three large genomewide association studies identified an as-sociation between single-nucleotide polymorphism (SNP) variation at 15q2415q25.1and susceptibility to lung cancer. The region of the SNP variation was recently

    linked to lung carcinogenesis and includes two genes encoding subunits of thenicotinic acetylcholine receptor alpha, which is regulated by nicotine exposure.19-22

    Lung-cancer susceptibility and risk also increase with reduced DNA repair ca-pacity (particularly when accompanied by exposure to tobacco smoke)23that re-sults, for example, from germ-line alterations in nucleotide excision repair genessuch as ERCC1.24Increased expression of DNA synthesis and repair genes, includ-ing RRM1(the regulatory subunit of ribonucleotide reductase) and ERCC1, in nonsmall-cell lung cancer correlates with a better prognosis overall but no benefitfrom platinum-based chemotherapy.25,26Table 1presents gene abnormalities in-volved in the development of different histologic types of lung cancer.

    The New England Journal of Medicine

    Downloaded from nejm.org on November 18, 2014. For personal use only. No other uses without permission.

    Copyright 2008 Massachusetts Medical Society. All rights reserved.

  • 8/10/2019 Molecular Origin Lung Cancer

    2/14

    T h e n e w e n g l a n d j o u r n a l o f medicine

    n engl j med 359;13 www.nejm.org september 25, 20081368

    Normal Precancer Early-stage cancer Advanced cancer

    Smoking-related

    Not smoking-related

    Molecularcellular

    features

    Treatmentapproaches

    Commongerm-line

    geneticvariations

    Prevention

    Cancer risk

    Host susceptibility

    Tobacco smoke

    Unknown factors

    Clonal patch

    Tissue injury

    Genetic,epigenetic

    changes

    Clonal patches

    Dysregulatedpathways,

    proliferation,apoptosis

    Invasion, angiogenesis

    Definitive local therapy with orwithout adjuvant therapy

    Prognostic, predictive

    Metastatic spread

    Systemic therapy with orwithout radiation therapy

    PredictiveRoles formolecularmarkers

    :i

    l ll

    l

    I

    l l l i l

    Figure 1.Molecular Evolution of Lung Cancer.

    Environmental factors, such as tobacco smoke, and genetic susceptibility interact to influence carcinogenesis. Factors that are unrelated

    to smoking including genetic, hormonal, and viral (e.g., human papillomavirus) factors have been suggested.2

    Tissue injury (e.g.,from tobacco smoke, reflected in the discolored smoking-related lungs) initially occurs in the form of genetic and epigenetic changes (e.g.,mutations, loss of heterozygosity, and promoter methylation) and global transcriptome changes (e.g., inflammation and apoptosis path-ways). These changes can persist long term3,4and eventually lead to aberrant pathway activation and cellular function (e.g., dysregulatedproliferation and apoptosis) to produce premalignant changes, including dysplasia and clonal patches. Additional changes can result inangiogenesis, invasion and early-stage cancer, and advanced cancer and metastasis.5Many molecular changes in earliest-stage canceralso occur in advanced disease.6,7Premalignant patches contain clones and subclones (inset), which can involve loss of heterozygosity,microsatellite instability, and mutations (e.g., in p53and epidermal growth factor receptor [EGFR]). Lung cancers unrelated and related tosmoking have strikingly different molecular profiles, including those of mutations in p53, KRAS, EGFR,and HER2. Smoking-related patchesand primary cancers (usually squamous-cell carcinoma and small-cell lung cancer) most of ten develop in the central airway.4,8Most tumorsthat are not related to smoking are adenocarcinomas and develop in the peripheral airways. Molecular markers can signify risk (in people with-out cancer), prognosis (outcome independent of treatment), and sensitivity to treatment through predictive markers. Such stage-specific mark-ers can span the course of disease from its early stages through its late stages. They also can help define mechanisms of resistance to therapy.

    The New England Journal of Medicine

    Downloaded from nejm.org on November 18, 2014. For personal use only. No other uses without permission.

    Copyright 2008 Massachusetts Medical Society. All rights reserved.

  • 8/10/2019 Molecular Origin Lung Cancer

    3/14

    Molecular Origins of Cancer

    n engl j med 359;13 www.nejm.org september 25, 2008 1369

    Clonal Evolution

    Changes in certain genes (e.g., proinflammatoryinterleukin-8 [IL8] and some DNA-repair genes)occur in nonmalignant lung tissue of smokersand patients with lung cancer, a finding consis-tent with diffuse tissue injury.3,27-29These chang-es probably precede epithelial clonal evolution, animportant element of the molecular origins oflung and other cancers (Fig. 1). Patches of clon-

    ally related cells, or clonal patches containing40,000 to 360,000 cells, have been mapped in thelung.30The size and number of subclones in aclonal patch may contribute to the cancer risk.31Early events in the development of nonsmall-cell lung cancer include loss of heterozygosity atchromosomal region 3p21.3 (site of RASSF1A, amember of the Ras association domain family,and FUS1), 3p14.2 (FHIT,a fragile histidine triad

    Table 1.Genetic Abnormalities Specific in the Lung to NonSmall-Cell Lung Cancer and Small-Cell Lung Cancer.*

    Abnormality NonSmall-Cell Lung Cancer Small-Cell Lung Cancer

    Squamous-CellCarcinoma Adenocarcinoma

    Precursor

    Lesion Known (dysplasia) Probable (atypical adenomatous hyperplasia) Possible (neuroendocrine field)

    Genetic change p53mutation KRASmutation (atypical adenomatous hyperplasiain smokers), EGFRkinase domain mutation(in nonsmokers)

    Overexpression of c-MET

    Cancer

    KRASmutation Very rare 10 to 30% Very rare

    BRAFmutation 3% 2% Very rare

    EGFR

    Kinase domain mutation Very rare 10 to 40% Very rare

    Amplification 30% 15% Very rare

    Variant III mutation 5% Very rare Very rare

    HER2

    Kinase domain mutation Very rare 4% Very rare

    Amplification 2% 6% Not known

    ALKfusion Very rare 7% Not known

    MET

    Mutation 12% 14% 13%

    Amplification 21% 20% Not known

    TITF-1amplification 15% 15% Very rare

    p53mutation 60 to 70% 50 to 70% 75%

    LKB1mutation 19% 34% Very rare

    PIK3CA

    Mutation 2% 2% Very rareAmplification 33% 6% 4%

    * Nonsmall-cell lung cancer includes squamous-cell carcinoma and adenocarcinoma. Neuroendocrine fields have been detected only in tissue surrounding tumors and have been characterized by extremely high rates of allelic

    lossand by c-MET overexpression (Salgia R: personal communication). Variations are based in part on smoking profiles. The percentages include increased gene copy numbers from amplification or polysomy and represent percentages from resected cancers.

    The percentages are higher in primary tumors from patients with metastatic disease. Increased copy numbers have been reported insquamous dysplastic lesions but not in adenocarcinoma precursors.

    Genomic EGFRvariant III mutations have been detected only in lung squamous-cell carcinoma, and these tumors are sensitive preclinicallyto irreversible EGFR tyrosine kinase inhibitors. The incidence of 5% is substantially lower than that of 30 to 40% for the detection insquamous-cell carcinoma or adenocarcinoma by immunohistochemical analysis or other techniques.

    The anaplastic lymphoma kinase (ALK) fusion gene (involving chromosome 2p), consisting of parts of EML4andALK,is transforming infibroblasts and occurs in adenocarcinoma but not in other types of nonsmall-cell lung cancer or other nonlung cancers.

    The New England Journal of Medicine

    Downloaded from nejm.org on November 18, 2014. For personal use only. No other uses without permission.

    Copyright 2008 Massachusetts Medical Society. All rights reserved.

  • 8/10/2019 Molecular Origin Lung Cancer

    4/14

    T h e n e w e n g l a n d j o u r n a l o f medicine

    n engl j med 359;13 www.nejm.org september 25, 20081370

    gene), 9p21 (p16), and 17p13 (p53).32 All thesegenes are tumor-suppressor genes. Loss-of-hetero-zygosity patterns in squamous-cell carcinomaand adenocarcinoma differ (e.g., chromosome 3pdeletions are much more extensive in squamous-cell carcinoma). Mutations in the EGFR kinase

    domain occur early in the development of adeno-carcinoma that is generally unrelated to smok-ing, and KRASmutations occur early in the devel-opment of smoking-related adenocarcinoma.33,34Clonal patches with methylation of promoter re-gions of genes (epigenetic changes),p53mutation,EGFRmutation, c-Mycamplification, loss of hetero-zygosity, and microsatellite instability can occurin normal tissue surrounding nonsmall-cell lungtumors5,30,32,34,35and may be associated with a

    greater risk of recurrence and second primarytumors. These findings suggest that in the futuremolecular analyses of surgical margins may helpidentify patients most likely to benefit from adju-vant therapy.

    Methylated genes in premalignant squamous-

    cell lung lesions (e.g., metaplasia and dysplasia) arep16and FHIT(frequently and very early) and O-6-methylguanine-DNA methyltransferase (MGMT),death-associated protein kinase (DAPK), andRASSF1A(less frequently or rarely and in advancedprecancers).35-41The early methylation of p16inthe development of squamous-cell lung cancer(e.g., in normal lung tissue in approximately50% of smokers)36exemplifies differences withthat in the development of adenocarcinoma, in

    Hypoxia

    Ligand bindingand dimerization

    EGFR

    LKB1

    AMPK

    TSC2

    HIF-1 mTOR

    Akt

    PI3K

    Mek

    Raf

    Ras

    Gene transcription, cellular effects

    Proliferation Invasion Metastasis Resistance to apoptosis Angiogenesis

    Other receptor tyrosinekinases (e.g., IGF-1R, c-Met)

    VEGF

    Figure 2.Epidermal Growth Factor Receptor (EGFR) Cell-Signaling Pathways.

    EGFR activates several major downstream signaling pathways, including RasRafMek and the pathway consistingof phosphoinositide 3-kinase (PI3K), Akt, and mammalian target of rapamycin (mTOR), which in turn may have aneffect on proliferation, survival, invasiveness, metastatic spread, and tumor angiogenesis through pathways that areeither dependent on or independent of the hypoxia inducible factor (HIF). These pathways also may be modulatedby other receptor tyrosine kinases, such as insulin-like growth factor 1 receptor (IGF-1R) and cMET, and by the

    LKB1amp-activated protein kinase (AMPK) pathway, which is involved in energy sensing and cellular stress. Mostof these functions depend on signaling through the kinase domain. However, kinase-independent functions, suchas maintaining glucose transport, have been reported.46TSC2 denotes tuberous sclerosis complex 2, and VEGF vas-cular endothelial growth factor.

    The New England Journal of Medicine

    Downloaded from nejm.org on November 18, 2014. For personal use only. No other uses without permission.

    Copyright 2008 Massachusetts Medical Society. All rights reserved.

  • 8/10/2019 Molecular Origin Lung Cancer

    5/14

    Molecular Origins of Cancer

    n engl j med 359;13 www.nejm.org september 25, 2008 1371

    which p16 methylation occurs very rarely andonly late in precursors (e.g., high-grade atypicaladenomatous hyperplasia).37Methylation markersin sputum are associated with the risk of lungcancer (e.g., methylated p16)38and the recurrenceof lung cancer (methylated ASC-TMS1,39 alsocalled PYCARD). Recent data show that promoter

    methylation of various genes, including p16 instage I nonsmall-cell lung cancer, is associatedwith recurrence after resection.40,41Agents thatreverse epigenetic changes have shown promisein a mouse model of lung carcinogenesis and arebeing tested in humans with lung cancer.42

    Bronchoalveolar stem cells, which may beprecursors of lung adenocarcinoma, were identi-fied recently in studies in mice.43The KRAS, Pten,phosphoinositide 3-kinase (PI3K), and cyclin-dependent kinase pathways have been implicatedin the proliferation of these stem cells.44,45The

    potential role of bronchoalveolar stem cells andother tumorigenic stem-cell populations in thedevelopment and prognosis of human lung can-cer and its resistance to drugs is an importantarea of future investigation.

    Molecular Evolution

    EGFR Family

    EGFR regulates important tumorigenic process-es, including proliferation, apoptosis, angiogene-sis, and invasion (Fig. 2), and, along with itsligands, is frequently overexpressed in the devel-opment and progression of nonsmall-cell lungcancer.2,5,34,46Clinical trials of the EGFR tyrosinekinase inhibitor erlotinib for second-line or third-line treatment of such tumors and of the mono-clonal antibody against EGFR, cetuximab (com-bined with chemotherapy), for treatment ofpreviously untreated, advanced disease47,48vali-dated EGFR as a molecular target for therapy.EGFRmutations that were discovered during clini-cal trials led to extensive studies of the roles of

    these mutations and EGFRamplification in thepathogenesis of the disease and its prognosisand sensitivity to treatment.

    Several groups of investigators independentlyidentified somatic mutations in the kinase do-main of EGFRin lung adenocarcinoma in approxi-mately 10% of specimens from patients in theUnited States and in 30 to 50% of specimensfrom patients in Asia49(Fig. 3). The mutationsoccur with increased frequency in women and

    nonsmokers and are tightly associated with sensi-tivity to the EGFR tyrosine kinase inhibitors gefi-

    tinib and erlotinib and so appear to explain mostof the dramatic responses to these agents.50-52More than 80% of these mutations in lung can-cer involve in-frame deletions within exon 19 orthe L858R mutant within exon 21. EGFRmuta-tions are associated with an improved prognosisin nonsmall-cell lung cancer, even when treatedwith cytotoxic chemotherapy.53,54EGFRamplifi-cation is detected in dysplasia (especially of a highgrade), which is associated with lung-cancer risk

    Extracellulardomain

    Transmembranedomain

    Ligand-bindingpocket

    Intracellulardomain

    EGFRvariant III del(del exons 27)Squamous-cell

    carcinoma

    Catalyticdomain

    Exon 19 delAdenocarcinoma

    L858R(exon 21 del)

    Adenocarcinoma

    ATP-binding pocket

    T790MAdenocarcinoma

    PhosphorylationP

    :i

    l ll

    l

    I

    l i ii l

    i ll i

    Figure 3.Effect of Deletions and Mutations in the Epidermal Growth FactorReceptor Gene (EGFR) on Disease Development and Drug Targeting.

    Ligand binding to the EGFR extracellular domain results in receptor ho-modimerization and tyrosine phosphorylation, with the phosphate derivedfrom ATP bound within the kinase catalytic domain. EGFRmutations havetransforming potential in preclinical lung models and can occur early in hu-

    man lung carcinogenesis. EGFRpoint mutations (e.g., L858R) and exon 19deletions, which occur predominantly in adenocarcinoma of the lung, arelocated within the catalytic domain and result in constitutive EGFR activa-tion. These mutations are associated with increased sensitivity to EGFR ty-rosine kinase inhibitors, such as erlotinib and gefitinib. In contrast, muta-tions in T790M (an amino acid located within the ATP binding site of theEGFRkinase domain) are associated with acquired resistance to these drugs.EGFRvariant III mutant deletions occur in the extracellular domain and areassociated with squamous-cell cancer.

    The New England Journal of Medicine

    Downloaded from nejm.org on November 18, 2014. For personal use only. No other uses without permission.

    Copyright 2008 Massachusetts Medical Society. All rights reserved.

  • 8/10/2019 Molecular Origin Lung Cancer

    6/14

    T h e n e w e n g l a n d j o u r n a l o f medicine

    n engl j med 359;13 www.nejm.org september 25, 20081372

    when detected in the sputum of smokers, and isassociated with a poor prognosis but also withsensitivity to EGFR inhibitors.55,56The epidemio-logic links highlight three factors whether thepatient is a nonsmoker, Asian, and female that are associated independently and collective-ly with an improved response to EGFR tyrosine

    kinase inhibitors. However, erlotinib appearsto prolong survival in virtually all subgroups ofpatients with nonsmall-cell lung cancer.48Thereare major differences in clinical, pathological,sex-related, and molecular factors between smok-ers and lifelong nonsmokers in whom lung can-cer develops (Fig. 1).

    The vast majority of patients who have aninitial response to erlotinib and gefitinib even-tually have a relapse.49,57,58Recent studies haveidentified EGFRT790M mutations (in exon 20) intumors before drug treatment59 and in tumors

    of patients who had a relapse after therapy withstandard reversible EGFR tyrosine kinase inhibi-tors.57The binding kinetics of the mutant EGFRappear to be altered by the T790M mutation(Fig. 3). Irreversible EGFR inhibitors suppressT790M-mutant tumor cells in vitro and are prom-ising treatments for T790M-mutant tumors.60Amplification of the met proto-oncogene (MET),another major mechanism of acquired resistanceto EGFR tyrosine kinase inhibitors, marks a poorprognosis.61-63Other proposed resistance mech-anisms include activation of other receptor tyro-sine kinases, such as insulin-like growth factor 1receptor (which can bypass EGFR to activatecritical downstream signaling pathways [Fig. 2]),KRASmutations, and the epithelial-to-mesenchy-mal transition.58,64,65 The epithelial-to-mesen-chymal transition is a program of cell develop-ment involving loss of cell adhesion, repressedE-cadherin expression, and increased cell mobility.

    Preclinical and clinical data suggest that EGFRmutations are early events in the development ofnonsmall-cell lung cancer.66-70EGFRmutations,

    including those involving exons 18, 19, and 20and L858R, can transform fibroblasts and lungepithelial cells.67Furthermore, in transgenic micewith lung-specific expression of exon 19 deletionor the L858R mutation, atypical adenomatoushyperplasia, which is considered to be a precur-sor lesion of peripheral adenocarcinoma, wasfollowed by lesions resembling bronchioalveolarcarcinoma at 5 to 6 weeks of age and invasiveadenocarcinomas at 8 to 10 weeks.68Deinduction

    of mutant EGFRexpression led to regression oftumors, suggesting the need for persistent mu-tant EGFRactivity for continued tumor survival.Lung tumors also developed in transgenic micewith lung-specific expression of EGFRvariant IIImutation (in-frame deletion of exons 27 fromthe extracellular domain) (Fig. 3).13Mutations of

    the region that encodes the tyrosine kinase do-main of EGFRhave been detected in specimensof atypical adenomatous hyperplasia from Asianpatients with no history of smoking.71These mu-tations also occur in normal epithelium withinand adjacent to tumors with EGFRtyrosine kinasemutations (a localized-field effect, possibly reflect-ing stem-cell expansion) and before EGFRampli-fication, a change associated with tumor progres-sion and metastasis.72

    HER2mutations and amplification have beenidentified in patients with lung adenocarcinoma.

    The frequency of such mutations is less than 5%,and the frequency of such amplification is 5 to10%. HER2kinase domain mutations (in-frameinsertions in exon 20) and EGFRkinase domainmutations have similar associations with femalesex, nonsmoking status, and Asian backgroundin patients with adenocarcinoma.73HER2amplifi-cation is associated with sensitivity to inhibitorsof the EGFR tyrosine kinase74; HER2mutations areassociated with resistance to such inhibitors butalso with sensitivity to HER2-targeted therapy.75HER3kinase domain mutations have not beendetected in patients with nonsmall-cell lungcancer.76Mutations in the HER4kinase domainwere found in 2 to 3% of Asian patients withthis disease, with a possible association withmale sex and smoking.77

    RasRafMek

    The RasRafMek pathway is involved in signal-ing downstream from EGFR and in other path-ways leading to the growth of cancer cells andtumor progression (Fig. 2). Activating KRASmu-

    tations are limited to nonsmall-cell lung cancer(predominantly adenocarcinomas), virtually mu-tually exclusive of mutations in the EGFR andHER2kinase domains, and associated with resis-tance to EGFR inhibitors (tyrosine kinase inhibi-tors and cetuximab) and chemotherapy.58,78,79Most KRAS mutations in lung adenocarcinomaare smoking-related GT transversions (substitu-tions of a purine for a pyrimidine) and affectexon 12 (in 90% of patients) or exon 13.2,79A dis-

    The New England Journal of Medicine

    Downloaded from nejm.org on November 18, 2014. For personal use only. No other uses without permission.

    Copyright 2008 Massachusetts Medical Society. All rights reserved.

  • 8/10/2019 Molecular Origin Lung Cancer

    7/14

    Molecular Origins of Cancer

    n engl j med 359;13 www.nejm.org september 25, 2008 1373

    tinct KRASmutational profile consisting of GAtransition mutations was recently detected inpatients with adenocarcinoma who had neversmoked; its functional significance is unclear.79Transversions (smokers) and transitions (non-smokers) also have been reported for p53muta-tions in lung adenocarcinoma.2KRASmutations

    appear to be an early event (e.g., detectable in thepreinvasive lesions of atypical adenomatous hy-perplasia and bronchoalveolar carcinoma33) thatprecedes smoking-related lung adenocarcinoma.They generally mark a poor prognosis. Furtherevidence supporting this genes role in the patho-genesis of lung cancer comes from transgenicmice bearing a mutated KRASand in which multi-focal atypical adenomatous hyperplasia and ade-nocarcinoma develop.80 MET activation occursearly in KRAS-induced carcinogenesis in this mod-el.81BRAFmutations have also been detected in

    nonsmall-cell lung cancer9and may be an earlyevent in lung tumorigenesis.82

    PI3KAktmTOR

    The pathway consisting of PI3K, Akt, and mam-malian target of rapamycin (mTOR), which isdownstream of EGFR, is activated early in lungcarcinogenesis.83 Akt is also overexpressed inbronchial dysplasia. Inhibition of Akt can induceapoptosis of human premalignant and malignantlung cells and prevent lung carcinogenesis in ananimal model. An mTOR inhibitor can block ma-lignant progression of atypical adenomatous hyper-plasia lesions in the KRasmouse model.84SincemTOR drives tumorigenesis in part through mac-rophages, a prominent component of the tumormicroenvironment, the antitumor effect of mTORinhibition requires the tumor microenvironment.There is mutation or amplification of PIK3CA,which encodes the PI3K catalytic subunit, in a sub-group of nonsmall-cell lung tumors, especiallysquamous-cell carcinoma, in association withincreased PI3K activity and Akt expression.85

    LKB1

    LKB1(also called STK11) is frequently mutated innonsmall-cell lung tumors and is thought to actas a tumor-suppressor gene through interactionswith p53and CDC42,modulating the activity ofAMPK (a multifunctional protein kinase) andother possible mechanisms that are just begin-ning to be studied.86,87LKB1is thought to func-tion in early tumorigenesis, subsequent differen-

    tiation, and the development of metastases.88Results in transgenic KRas-mutant mice in whichLKB1was inactivated suggest that the gene playsa role in the differentiation and invasive behaviorof such tumors.87The presence of LKB1mutationsalone (i.e., without KRasmutations) was not asso-ciated with the development of lung cancer in

    mice. Low levels of LKB1 protein were associatedwith high grades of dysplasia in atypical adeno-matous hyperplasia lesions, suggesting that LKB1has an early role in the development of premalig-nant lesions in the lung.89LKB1mutations (includ-ing point mutations and deletions) were found in34% of adenocarcinomas and 19% of squamous-cell carcinomas from 144 human specimens ofnonsmall-cell lung cancer.87However, much low-er rates of LKB1mutation (

  • 8/10/2019 Molecular Origin Lung Cancer

    8/14

    T h e n e w e n g l a n d j o u r n a l o f medicine

    n engl j med 359;13 www.nejm.org september 25, 20081374

    of bronchial dysplasia from low grade to highgrade.95Bronchial hyperplasia, metaplasia, andcarcinoma in situ are associated with increasedmicrovessel density, and a distinctive patternknown as angiogenic squamous dysplasia canoccur.96Factors associated with increased tumorangiogenesis correlate with the development and

    prognosis of lung cancer.97-99Circulating VEGFlevels may predict the clinical benefit of VEGFinhibitors in patients with this disease. Many an-giogenic factors are regulated at least in partthrough the hypoxia-regulated pathways, such ashypoxia-induced factor (HIF) 1 and 2.100,101Inaddition to hypoxia, VEGF and other angiogenicfactors are also regulated by EGFR through HIF-dependent and independent mechanisms102andby oncogenes such as KRASand p53. VEGF hasrecently been validated as a therapeutic target onthe basis of the results of a phase 3 trial, which

    led the Food and Drug Administration (FDA) toapprove the VEGF monoclonal antibody bevaci-zumab in combination with standard chemother-apy for previously untreated, advanced nonsmall-cell lung cancer.103

    Interactions between the VEGF and EGFRpathways and an association between acquiredresistance to EGFR blockade and increased VEGFexpression in preclinical models104 led to thehypothesis that dual blockade of VEGF and EGFRmight be more effective than either approachalone. Randomized phase 2 trials of dual inhibi-tion with bevacizumab plus erlotinib105 or theVEGF receptorEGFR tyrosine kinase inhibitorvandetanib (combined with chemotherapy)106,107had promising results. Phase 3 testing of bothapproaches in patients with platinum-resistantdisease is ongoing.

    molecular profiling

    technical advances

    Molecular profiling, including the profiling of

    genes and proteins, to guide treatment may im-prove the clinical outcome in patients with nonsmall-cell lung cancer (Fig. 1). Progress in theidentification of markers, mutations, and genomicsignatures far outstrips the modest improvementin treatments that are based on these molecularadvances. Formidable obstacles to developing ef-fective markers include tumor heterogeneity, thehighly complex interplay between the environ-ment and host and the complexity, multiplicity,

    and redundancy of tumor-cell signaling networksinvolving genetic, epigenetic, and microenviron-mental effects. Emerging high-throughput tech-niques for assessing genomic DNA, messengerRNA (mRNA), microRNA, methylation, and pro-tein or phosphoprotein signaling networks shouldhelp address these obstacles (Fig. 4). The Cancer

    Genome Atlas is a large-scale project designedto provide a comprehensive profile of humantumors according to their gene mutations, alter-ations in gene copy number, and epigeneticchanges. Squamous-cell carcinoma of the lungwill be one of the f irst tumors profi led by thisatlas.

    Gene Profiling

    Tumor molecular heterogeneity is a major reasonthat patients with nonsmall-cell lung cancer witha similar clinical stage and tumor histology can

    have dramatically different clinical outcomes andresponses to treatment. Microarray techniquesthat profile the expressions of tens of thousandsof genes simultaneously can measure this tumorheterogeneity at a global level. Gene-expressionprofiles that are associated with subtypes of nonsmall-cell lung cancer108,109 and with reducedrecurrence-free or overall survival of patientshave been identified.110-113Combined clinical andmolecular information provides better indicationsof cancer risk114and prognosis.115

    In a recent analysis of 672 invasion-associatedgenes from 125 frozen specimens of early-stagetumors,111microarray and reverse-transcriptasepolymerase-chain-reaction (RT-PCR) analyses iden-tified a molecular signature of five genes as anindependent predictor of relapse-free and overallsurvival. In two validation cohorts, another re-cently developed gene-expression profile (meta-gene) predicted clinical outcome with an accuracyof 72% and 79% (greater than that for tumorstage, tumor diameter, nodal status, or otherclinical measures) and predicted the outcome in

    patients with stage IA tumors.112

    Randomized,controlled trials will need to validate these sig-natures and establish whether the patients withstage IA tumors who were identified as being athigh risk will benefit from adjuvant therapy. Onesuch phase 3 trial, coordinated by the Cancerand Leukemia Group B, is approved and underfinal review. It will evaluate a large predictive setof metagenes (or subgroups of gene-expressionprofiles consisting of 25 to 200 genes) in patients

    The New England Journal of Medicine

    Downloaded from nejm.org on November 18, 2014. For personal use only. No other uses without permission.

    Copyright 2008 Massachusetts Medical Society. All rights reserved.

  • 8/10/2019 Molecular Origin Lung Cancer

    9/14

    Molecular Origins of Cancer

    n engl j med 359;13 www.nejm.org september 25, 2008 1375

    with stage IA tumors who are undergoing adju-vant chemotherapy.

    For the majority of patients with advanced ormetastatic nonsmall-cell lung cancer, the most

    important potential effect of molecular markersis likely to be in predicting the response to spe-cific therapies with the goal of personalizingtreatment (Fig. 4). Many exciting potential pre-dictive markers have been developed in vitro andneed validation in tumor samples and clinicaltrials.113For example, gene-expression signatureshave been developed for cisplatin and pemetrexedon the basis of in vitro sensitivity; the cisplatinin vitro signature predicted the likelihood of

    response. Recently developed in vitro profilespredicting the sensitivity of tumors to EGFR in-hibitors and other therapies have yet to be as-sessed clinically.113

    MicroRNA has recently emerged as an impor-tant regulator of gene expression. High-through-put analyses have shown that microRNA expres-sion is commonly deregulated in lung and othercancers.116,117Using real-time RT-PCR, investi-gators recently identified a five-microRNA sig-nature that is associated with treatment out-come.116 Loss of microRNA-128b, a putativeregulator of EGFR that is located on chromo-some 3p, has been shown to correlate with the

    Host

    Tumor, local environment

    Germ-line genetic profiling(e.g., SNP arrays)

    Blood-based profiling

    ProteomicsCytokine, angiogenic factorsCirculating DNA or RNACirculating cells (e.g., tumor,endothelial, lymphocytes)

    Genetic or epigenetic

    MutationDeletionAmplificationTranslocationMethylation

    Gene expression

    Expression arraysMicroRNA profilingPCR-based approaches

    Proteins

    IHCProteomics

    Personalizedmedicine

    Interactions involving the host, tumor,and therapy

    Tumor sensitivity to therapy

    :i

    l ll

    l

    I

    l l ili

    l li

    Figure 4.Molecular-Profiling Approaches to the Development of Personalized Therapy.

    Host profiling involves innate characteristics of the cancer patient. All markers that are involved in profiling lung cancer can apply to thetumor or its local environment. Predictive markers identify groups of patients who are likely to have increased sensitivity or resistance toa given therapy, a critical step in personalizing treatment. It has been traditional to assess individual genetic or protein prognostic orpredictive markers (e.g., HER2 for breast cancer), but emerging techniques permit global analyses of the genomic, gene-expression, epi-

    genetic, and protein profiles of the host (innate), including markers in blood and in tumor or nonmalignant lung tissue. These methodsinclude single-nucleotide polymorphism (SNP) arrays to assess genomic alterations, bisulfite sequencing, and methylation-specific poly-merase chain reaction (PCR) to assess epigenetic changes, microarrays for assessing gene expression or microRNA levels, and proteom-ic methods (such as mass spectroscopy, reverse-phase protein arrays, and multiplex beads) to assess intracellular signaling in tumor tis-sue and cytokines and angiogenic factors in blood. Blood-based profiling includes markers derived from the host (e.g., lymphocytes)and the tumor and local environment (e.g., circulating tumor cells and tumor-derived cytokines) (red arrows). IHC denotes immunohis-tochemical analysis.

    The New England Journal of Medicine

    Downloaded from nejm.org on November 18, 2014. For personal use only. No other uses without permission.

    Copyright 2008 Massachusetts Medical Society. All rights reserved.

  • 8/10/2019 Molecular Origin Lung Cancer

    10/14

    T h e n e w e n g l a n d j o u r n a l o f medicine

    n engl j med 359;13 www.nejm.org september 25, 20081376

    response to EGFR inhibition in patients withlung cancer.117

    Studies suggest that information about tumor-specific genetic and epigenetic changes also maybe obtained from the blood of patients with lungcancer. Circulating DNA can be detected in theplasma and serum of such patients, and levels of

    this DNA are associated with a poor progno-sis.118,119Tumor-specific DNA alterations (suchas loss of heterozygosity), promoter methylation,and KRASand EGFRmutations have also beendetected in the blood of patients with lung can-cer.120-122New techniques for capturing circulat-ing tumor cells allow the detection of EGFR-activating mutations and the drug-resistanceallele T790M. Such techniques appear to be moresensitive than those for capturing circulatingDNA. Furthermore, a decline in the number ofcirculating tumor cells was associated with tu-

    mor response on radiography.123These studiessuggest that blood profiling may provide usefulinformation about genetic changes in tumorsthat could ultimately help detect lung cancer andguide therapy.

    Protein Profiling

    Profiling of genomic and mRNA expression pro-vides an incomplete picture of the heterogeneityof nonsmall-cell lung cancer. Levels of mRNAdo not always correlate with protein levels and donot provide information on proteinprotein inter-actions or post-translational modifications suchas phosphorylation that may be critical for regu-lating protein activity.124Furthermore, most tar-geted therapeutic agents are designed to inhibitthe activity of proteins such as tyrosine kinases.Therefore, protein-based profiling is likely to beessential in understanding the complexity of pro-tein signaling networks and developing molecu-lar signatures that predict a response to therapy.

    Immunohistochemical analysis remains themost widely applied method for assessing indi-

    vidual proteins and may be useful for estimatingprognosis and predicting the response to ther-apy.25,26 Emerging high-throughput proteomictechniques, such as mass spectrometry and pro-tein microarrays, have the potential to view sig-nal transduction networks more globally than ispossible with immunohistochemical analysis.Such techniques are feasible in small amounts oftumor tissue.125Proteomic signatures for prog-

    nosis and predicting the response to chemother-apy or EGFR inhibitors have been developed intumors and cell lines.126,127

    Proteomic profiling from blood is also understudy, allowing repeated measurements duringtreatment without the need for tumor tissue.Serum mass spectrometry profiles can distin-

    guish patients with nonsmall-cell lung cancerfrom normal controls124,128and patients with bet-ter outcomes from those with worse outcomesafter treatment with EGFR tyrosine kinase inhib-itors.129New techniques also permit the multi-plex analysis of dozens of cytokines and angio-genic factors in small amounts of serum orplasma. This approach is being used in develop-ing predictive markers in nonsmall-cell lungcancer. Although promising, blood- and tissue-based proteomic approaches remain investiga-tional and await prospective testing and valida-

    tion in large, randomized trials before they canbe applied clinically.

    Conclusions

    The molecular origins of lung cancer lie in com-plex interactions between the environment andhost genetic susceptibility. Lung cancer thenevolves through genetic and epigenetic changes,including deregulated signaling pathways, whichare potential targets for chemoprevention andtherapy. Emerging techniques for genomic, gene-expression, epigenetic, and proteomic profil-ing92,114,125,130,131could revolutionize clinical ap-proaches across the spectrum of lung-cancer typesand subtypes by identifying practical molecularmarkers of risk (in precancer), early detectionand prognosis (in early-stage cancer), and treat-ment sensitivity (in early-stage and advanced-stage cancer). Genomewide and other molecularassessments are helping elucidate germ-line vari-ations that may contribute to lung cancer risk,19-21prognosis,132and treatment sensitivity133,134and

    somatic genetic alterations that occur in lungadenocarcinomas14,50-52 and in high-risk lungtissue associated with tumors or in smokers.3,28,29Molecular targeted research has produced the re-cently FDA-approved EGFR and VEGF inhibitorserlotinib and bevacizumab, which have modestlyimproved the outcome in patients with nonsmall-cell lung cancer.48,103Molecular profiling of thetype described in this review has begun in clini-

    The New England Journal of Medicine

    Downloaded from nejm.org on November 18, 2014. For personal use only. No other uses without permission.

    Copyright 2008 Massachusetts Medical Society. All rights reserved.

  • 8/10/2019 Molecular Origin Lung Cancer

    11/14

    Molecular Origins of Cancer

    n engl j med 359;13 www.nejm.org september 25, 2008 1377

    cal trials112,135-137and promises to select patientswho are most likely to benefit from therapy andto guide the development of more effective agentsthat will personalize standard medicine for lungcancer.138

    Dr. Herbst reports receiving consulting fees and researchgrants from Bristol-Myers Squibb, ImClone, Genentech, Amgen,

    AstraZeneca, and OSI and lecture fees from Genentech; Dr.

    Heymach, serving on advisory boards for and receiving researchgrants from AstraZeneca, Pfizer, and GlaxoSmithKline and serv-ing on an advisory board for Genentech; and Dr. Lippman, servingon advisory boards for OSI and Genentech. No other potentialconflict of interest relevant to this article was reported.

    We thank Lauren A. Byers, Balvindar S. Johal, and Ignacio I.Wistuba for their careful comments and other contributions re-garding aspects of this work; and Bich N. Tran, Suzanne E. Davis,and Kendall Morse for their contributions to the preparation of

    the manuscript.

    References

    Jemal A, Siegel R, Ward E, et al. Can-1.cer statistics, 2008. CA Cancer J Clin 2008;58:71-96.

    Sun S, Schiller JH, Gazdar AF. Lung2.cancer in never smokers a differentdisease. Nat Rev Cancer 2007;7:778-90.

    Spira A, Beane J, Shah V, et al. Effects3.of cigarette smoke on the human airwayepithelial cell transcriptome. Proc NatlAcad Sci U S A 2004;101:10143-8.

    Mao L, Lee JS, Kurie JM, et al. Clonal4.genetic alterations in the lungs of current

    and former smokers. J Natl Cancer Inst1997;89:857-62.

    Sato M, Shames DS, Gazdar AF, Min-5.na JD. A translational view of the molecu-lar pathogenesis of lung cancer. J ThoracOncol 2007;2:327-43.

    Bianchi F, Hu J, Pelosi G, et al. Lung6.cancers detected by screening with spiralcomputed tomography have a malignantphenotype when analyzed by cDNA micro-array. Clin Cancer Res 2004;10:6023-8.

    Zudaire I, Lozano MD, Vazquez MF, et7.al. Molecular characterization of smallperipheral lung tumors based on the analy-sis of fine needle aspirates. Histol Histo-pathol 2008;23:33-40.

    Wistuba II, Berry J, Behrens C, et al.8.Molecular changes in the bronchial epi-thelium of patients with small cell lungcancer. Clin Cancer Res 2000;6:2604-10.

    Brose MS, Volpe P, Feldman M, et al.9.BRAF and RAS mutations in human lungcancer and melanoma. Cancer Res 2002;62:6997-7000.

    Kong-Beltran M, Seshagiri S, Zha J,10.et al. Somatic mutations lead to an onco-genic deletion of met in lung cancer. Can-cer Res 2006;66:283-9.

    Ma PC, Kijima T, Maulik G, et al. c-MET11.mutational analysis in small cell lung can-cer: novel juxtamembrane domain muta-

    tions regulating cytoskeletal functions.Cancer Res 2003;63:6272-81.Beau-Faller M, Ruppert AM, Voegeli12.

    AC, et al. MET gene copy number in non-small cell lung cancer: molecular analysisin a targeted tyrosine kinase inhibitor na-ve cohort. J Thorac Oncol 2008;3:331-9.

    Ji H, Zhao X, Yuza Y, et al. Epidermal13.growth factor receptor variant III muta-tions in lung tumorigenesis and sensitiv-ity to tyrosine kinase inhibitors. Proc NatlAcad Sci U S A 2006;103:7817-22.

    Soda M, Choi YL, Enomoto M, et al.14.

    Identification of the transforming EML4-ALK fusion gene in non-small-cell lungcancer. Nature 2007;448:561-6.

    Hwang SJ, Cheng LS, Lozano G, Amos15.CI, Gu X, Strong LC. Lung cancer risk ingermline p53 mutation carriers: associa-tion between an inherited cancer predis-position, cigarette smoking, and cancerrisk. Hum Genet 2003;113:238-43.

    Sanders BM, Jay M, Draper GJ, Rob-16.erts EM. Non-ocular cancer in relat ives ofretinoblastoma patients. Br J Cancer 1989;

    60:358-65.Bailey-Wilson JE, Amos CI, Pinney SM,17.

    et al. A major lung cancer susceptibilitylocus maps to chromosome 6q23-25. AmJ Hum Genet 2004;75:460-74.

    Bell DW, Gore I, Okimoto RA, et al.18.Inherited susceptibility to lung cancer maybe associated with the T790M drug resis-tance mutat ion in EGFR. Nat Genet 2005;37:1315-6.

    Amos CI, Wu X, Broderick P, et al.19.Genome-wide association scan of tag SNPsidentifies a susceptibility locus for lungcancer at 15q25.1. Nat Genet 2008;40:616-22.

    Hung RJ, McKay JD, Gaborieau V, et al.20.A susceptibility locus for lung cancer mapsto nicotinic acetylcholine receptor sub-unit genes on 15q25. Nature 2008;452:633-7.

    Thorgeirsson TE, Geller F, Sulem P,21.et al. A variant associated with nicotinedependence, lung cancer and peripheralarter ial disease. Nature 2008;452:638-42.

    Lam DC, Girard L, Ramirez R, et al.22.Expression of nicotinic acetylcholine recep-tor subunit genes in non-small-cell lungcancer reveals differences between smok-ers and nonsmokers. Cancer Res 2007;67:4638-47.

    Spitz MR, Wei Q, Dong Q, Amos CI,23.

    Wu X. Genetic susceptibility to lung can-cer: the role of DNA damage and repair.Cancer Epidemiol Biomarkers Prev 2003;12:689-98.

    Yu D, Zhang X, Liu J, et al. Character-24.ization of functional excision repair cross-complementation group 1 variants andtheir association with lung cancer riskand prognosis. Clin Cancer Res 2008;14:2878-86.

    Olaussen KA, Dunant A, Fouret P, et al.25.DNA repair by ERCC1 in nonsmall-celllung cancer and cisplatin-based adjuvant

    chemotherapy. N Engl J Med 2006;355:983-91.

    Zheng Z, Chen T, Li X, Haura E, Shar-26.ma A, Bepler G. DNA synthesis and repairgenes RRM1and ERCC1 in lung cancer.N Engl J Med 2007;356:800-8.

    Franklin WA, Gazdar AF, Haney J, et al.27.Widely dispersed p53 mutation in respira-tory epithelium: a novel mechanism forfield carcinogenesis. J Clin Invest 1997;100:2133-7. [Erratum, J Clin Invest 1997;100:2639.]

    Spira A, Beane JE, Shah V, et al. Air-28.way epithelial gene expression in the diag-nostic evaluation of smokers with suspectlung cancer. Nat Med 2007;13:361-6.

    Seike M, Yanaihara N, Bowman ED, et29.al. Use of a cytokine gene expression sig-nature in lung adenocarcinoma and thesurrounding tissue as a prognostic classi-fier. J Natl Cancer Inst 2007;99:1257-69.

    Park IW, Wistuba II, Maitra A, et al.30.Multiple clonal abnormalities in the bron-chial epithelium of patients with lung can-cer. J Natl Cancer Inst 1999;91:1863-8.

    Maley CC, Galipeau PC, Finley JC, et al.31.Genetic clonal diversity predicts progres-sion to esophageal adenocarcinoma. NatGenet 2006;38:468-73.

    Wistuba II, Mao L, Gazdar AF. Smok-32.ing molecular damage in bronchial epi-thelium. Oncogene 2002;21:7298-306.

    Westra WH. Early glandular neoplasia33.of the lung. Respir Res 2000;1:163-9.

    Tang X, Shigematsu H, Bekele BN, et al.34.EGFR tyrosine kinase domain mutationsare detected in histologically normal respi-ratory epithelium in lung cancer patients.Cancer Res 2005;65:7568-72.

    Guo M, House MG, Hooker C, et al.35.Promoter hypermethylation of resectedbronchial margins: a field defect of chang-es? Clin Cancer Res 2004;10:5131-6.

    Bhutani M, Pathak AK, Fan Y-H, et al.36.Oral epithelium as a surrogate tissue forassessing smoking-induced molecularalterations in the lungs. Cancer Prev Res2008;1:39-44.

    Licchesi JD, Westra WH, Hooker CM,37.Herman JG. Promoter hypermethylationof hallmark cancer genes in atypical ade-nomatous hyperplasia of the lung. ClinCancer Res 2008;14:2570-8.

    Belinsky SA, Liechty KC, Gentry FD,38.et al. Promoter hypermethylation of mul-tiple genes in sputum precedes lung can-

    The New England Journal of Medicine

    Downloaded from nejm.org on November 18, 2014. For personal use only. No other uses without permission.

    Copyright 2008 Massachusetts Medical Society. All rights reserved.

  • 8/10/2019 Molecular Origin Lung Cancer

    12/14

    T h e n e w e n g l a n d j o u r n a l o f medicine

    n engl j med 359;13 www.nejm.org september 25, 20081378

    cer incidence in a high-risk cohort. Can-cer Res 2006;66:3338-44.

    Machida EO, Brock MV, Hooker CM,39.et al. Hypermethylat ion of ASC/TMS1 is asputum marker for late-stage lung cancer.Cancer Res 2006;66:6210-8. [Erratum, Can-cer Res 2007;67:427.]

    Brock MV, Hooker CM, Ota-Machida E,40.et al. DNA methylation markers and early

    recurrence in stage I lung cancer. N EnglJ Med 2008;358:1118-28.Kim JS, Kim JW, Han J, Shim YM, Park41.

    J, Kim DH. Cohypermethylation of p16and FHIT promoters as a prognostic fac-tor of recurrence in surgically resectedstage I non-small cell lung cancer. CancerRes 2006;66:4049-54.

    Belinsky SA, Klinge DM, Stidley CA,42.et al. Inhibition of DNA methylation andhistone deacetylation prevents murinelung cancer. Cancer Res 2003;63:7089-93.

    Kim CF, Jackson EL, Woolfenden AE,43.et al. Identification of bronchioalveolarstem cells in normal lung and lung can-cer. Cell 2005;121:823-35.

    Yang Y, Iwanaga K, Raso MG, et al.44.Phosphatidylinositol 3-kinase mediatesbronchioalveolar stem cel l expansion inmouse models of oncogenic K-ras-inducedlung cancer. PLoS ONE 2008;3(5):e2220.

    Yanagi S, Kishimoto H, Kawahara K,45.et al. Pten controls lung morphogenesis,bronchioalveolar stem cells, and onset oflung adenocarcinomas in mice. J Clin In-vest 2007;117:2929-40.

    Weihua Z, Tsan R, Huang WC, et al.46.Survival of cancer cells is maintained byEGFR independent of its kinase activity.Cancer Cell 2008;13:385-93.

    Pirker R, Szczesna A, von Pawel J, et al.47.FLEX: a randomized, multicenter, phase III

    study of cetuximab in combination with cis-platin/vinorelbine (CV) versus CV alone inthe first-line treatment of patients with ad-vanced non-small cell lung cancer (NSCLC).J Clin Oncol 2008;26:Suppl:1006s. abstract.

    Shepherd FA, Rodrigues Pereira J,48.Ciuleanu T, et al. Erlotinib in previouslytreated nonsmall-cell lung cancer. N EnglJ Med 2005;353:123-32.

    Sequist LV, Bell DW, Lynch TJ, Haber49.DA. Molecular predictors of response toepidermal growth factor receptor antago-nists in non-small-cell lung cancer. J ClinOncol 2007;25:587-95.

    Lynch TJ, Bell DW, Sordella R, et al.50.Activating mutations in the epidermalgrowth factor receptor underlying respon-siveness of nonsmall-cell lung cancer togefitinib. N Engl J Med 2004;350:2129-39.

    Paez JG, Jnne PA, Lee JC, et al. EGFR51.mutations in lung cancer: correlation withclinical response to gefitinib therapy. Sci-ence 2004;304:1497-500.

    Pao W, Miller V, Zakowski M, et al.52.EGF receptor gene mutations are commonin lung cancers from never smokers andare associated with sensitivity of tumors

    to gefitinib and erlotinib. Proc Natl AcadSci U S A 2004;101:13306-11.

    Ciardiello F, Tortora G. EGFR antago-53.nists in cancer treatment. N Engl J Med2008;358:1160-74.

    Kim ES, Hirsh V, Mok T, et al. Gefi-54.tinib versus docetaxel in previously treat-ed non-small-cell lung cancer: a random-ized phase III trial (INTEREST). Lancet

    2008 (in press).Hirsch FR, Herbst RS, Olsen C, et al.55.Increased EGFR gene copy number de-tected by fluorescent in situ hybridizationpredicts outcome in non-small-cell lungcancer patients treated with cetuximaband chemotherapy. J Clin Oncol 2008;26:3351-7.

    Tsao M-S, Sakurada A, Cutz J-C, et al.56.Erlotinib in lung cancer molecular andclinical predictors of outcome. N Engl JMed 2005;353:133-44. [Erratum, N Engl JMed 2006;355:1746.]

    Pao W, Miller VA, Politi KA, et al. Ac-57.quired resistance of lung adenocarcino-mas to gefitinib or erlotinib is associated

    with a second mutation in the EGFR ki-nase domain. PLoS Med 2005;2(3):e73.

    Pao W, Wang TY, Riely GJ, et al. KRAS58.mutations and primary resistance of lungadenocarcinomas to gefitinib or erlotinib.PLoS Med 2005;2(1):e17.

    Inukai M, Toyooka S, Ito S, et al. Pres-59.ence of epidermal growth factor receptorgene T790M mutation as a minor clone innon-small cell lung cancer. Cancer Res2006;66:7854-8.

    Kwak EL, Sordella R, Bell DW, et al.60.Irreversible inhibitors of the EGF receptormay circumvent acquired resistance to ge-fit inib. Proc Natl Acad Sci U S A 2005;102:7665-70.

    Bean J, Brennan C, Shih JY, et al. MET61.amplification occurs with or withoutT790M mutations in EGFR mutant lungtumors with acquired resistance to gefi-tinib or erlotinib. Proc Natl Acad Sci U S A2007;104:20932-7.

    Engelman JA, Zejnullahu K, Mitsudo-62.mi T, et al. MET amplification leads togefit inib resistance in lung cancer by acti-vating ERBB3 signaling. Science 2007;316:1039-43.

    Cappuzzo F, Skokan M, Gajapathy S,63.et al. Effect of increased MET gene copynumber on survival of surgically resectednon-small cell lung cancer (NSCLC) pa-tients. J Clin Oncol 2008;26:Suppl:589s.abstract.

    Morgillo F, Kim WY, Kim ES, Ciar-64.diello F, Hong WK, Lee HY. Implication ofthe insulin-like growth factor-IR pathwayin the resistance of non-small cell lungcancer cells to treatment with gefitinib.Clin Cancer Res 2007;13:2795-803.

    Yauch RL, Januario T, Eberhard DA, et65.al. Epithelial versus mesenchymal pheno-type determines in vitro sensitivity andpredicts clinical activity of erlotinib in

    lung cancer patients. Clin Cancer Res2005;11:8686-98.

    Godin-Heymann N, Bryant I, Rivera66.MN, et al. Oncogenic activity of epidermalgrowth factor receptor kinase mutant al-leles is enhanced by the T790M drug re-sistance mutation. Cancer Res 2007;67:7319-26.

    Greulich H, Chen TH, Feng W, et al.67.

    Oncogenic transformation by inhibitor-sensitive and -resistant EGFR mutants.PLoS Med 2005;2(11):e313.

    Ji H, Li D, Chen L, et al. The impact of68.human EGFR kinase domain mutationson lung tumorigenesis and in vivo sensi-tivity to EGFR-targeted therapies. CancerCell 2006;9:485-95.

    Polit i K, Zakowski MF, Fan PD, Schon-69.feld EA, Pao W, Varmus HE. Lung adeno-carcinomas induced in mice by mutantEGF receptors found in human lung can-cers respond to a tyrosine kinase inhibi-tor or to down-regulation of the recep-tors. Genes Dev 2006;20:1496-510.

    Vikis H, Sato M, James M, et al. EGFR-70.

    T790M is a rare lung cancer susceptibilityallele with enhanced kinase activity. Can-cer Res 2007;67:4665-70.

    Sakuma Y, Matsukuma S, Yoshihara71.M, et al. Epidermal growth factor receptorgene mutations in atypical adenomatoushyperplasias of the lung. Mod Pathol 2007;20:967-73.

    Tang X, Varella-Garcia M, Xavier AC,72.et al. Epidermal growth factor receptorabnormalities in the pathogenesis andprogression of lung adenocarcinomas.Cancer Prev Res 2008;1:192-200.

    Shigematsu H, Takahashi T, Nomura73.M, et al. Somatic mutations of the HER2kinase domain in lung adenocarcinomas.

    Cancer Res 2005;65:1642-6.Cappuzzo F, Varella-Garcia M, Shige-74.

    matsu H, et al. Increased HER2 gene copynumber is associated with response to ge-fit inib therapy in epidermal growth factorreceptor-positive non-small-cell lung can-cer patients. J Clin Oncol 2005;23:5007-18.

    Wang SE, Narasanna A, Perez-Torres75.M, et al. HER2 kinase domain mutationresults in constitutive phosphorylationand activation of HER2 and EGFR and re-sistance to EGFR tyrosine kinase inhibi-tors. Cancer Cell 2006;10:25-38.

    Jeong EG, Soung YH, Lee JW, et al.76.ERBB3 kinase domain mutations are rarein lung, breast and colon carcinomas. IntJ Cancer 2006;119:2986-7.

    Soung YH, Lee JW, Kim SY, et al. So-77.matic mutations of the ERBB4 kinase do-main in human cancers. Int J Cancer 2006;118:1426-9.

    Eberhard DA, Johnson BE, Amler LC,78.et al. Mutations in the epidermal growthfactor receptor and in KRAS are predictiveand prognostic indicators in patients withnon-small-cell lung cancer treated withchemotherapy alone and in combination

    The New England Journal of Medicine

    Downloaded from nejm.org on November 18, 2014. For personal use only. No other uses without permission.

    Copyright 2008 Massachusetts Medical Society. All rights reserved.

  • 8/10/2019 Molecular Origin Lung Cancer

    13/14

    Molecular Origins of Cancer

    n engl j med 359;13 www.nejm.org september 25, 2008 1379

    with erlotinib. J Clin Oncol 2005;23:5900-9.

    Riely GJ, Kris MG, Rosenbaum D, et79.al. Frequency and distinctive spectrum ofKRAS mutations in never smokers withlung adenocarcinoma. Clin Cancer Res(in press).

    Johnson L, Mercer K, Greenbaum D,80.et al. Somatic activation of the K-ras onco-

    gene causes early onset lung cancer inmice. Nature 2001;410:1111-6.Yang Y, Wislez M, Fujimoto N, et al.81.

    A selective small molecule inhibitor ofc-Met, PHA-665752, reverses lung prema-lignancy induced by mutant K-ras. MolCancer Ther 2008;7:952-60.

    Dankort D, Filenova E, Collado M,82.Serrano M, Jones K, McMahon M. A newmouse model to explore the initiation,progression, and therapy of BRAFV600E-induced lung tumors. Genes Dev 2007;21:379-84.

    West KA, Linnoila IR, Belinsky SA,83.Harris CC, Dennis PA. Tobacco carcinogen-induced cellular transformation increases

    activation of the phosphatidylinositol 3-kinase/Akt pathway in vitro and in vivo.Cancer Res 2004;64:446-51.

    Wislez M, Spencer ML, Izzo JG, et al.84.Inhibition of mammalian target of ra-pamycin reverses alveolar epithelial neo-plasia induced by oncogenic K-ras. CancerRes 2005;65:3226-35.

    Yamamoto H, Shigematsu H, Nomura85.M, et al. PIK3CA mutations and gene copynumber in human lung cancers. CancerRes (in press).

    Zhang S, Schafer-Hales K, Khuri FR,86.Zhou W, Vertino PM, Marcus AI. The tumorsuppressor LKB1 regulates lung cancercell polarity by mediating cdc42 recruit-

    ment and activity. Cancer Res 2008;68:740-8.

    Ji H, Ramsey MR, Hayes DN, et al.87.LKB1 modulates lung cancer differentia-tion and metastasis. Nature 2007;448:807-10.

    Shah U, Sharpless NE, Hayes DN.88.LKB1 and lung cancer: more than the usu-al suspects. Cancer Res 2008;68:3562-5.

    Ghaffar H, Sahin F, Sanchez-Cepedes89.M, et al. LKB1 protein expression in theevolution of glandular neoplasia of thelung. Clin Cancer Res 2003;9:2998-3003.

    Matsumoto S, Iwakawa R, Takahashi90.K, et al. Prevalence and specificity ofLKB1 genetic alterations in lung cancers.Oncogene 2007;26:5911-8.

    Koivunen JP, Kim J, Lee J, et al. Muta-91.tions in the LKB1 tumour suppressor arefrequently detected in tumours from Cau-casian but not Asian lung cancer patients.Br J Cancer 2008;99:245-52.

    Weir BA, Woo MS, Getz G, et al. Char-92.acterizing the cancer genome in lung ad-enocarcinoma. Nature 2007;450:893-8.

    Kendall J, Liu Q, Bakleh A, et al. On-93.cogenic cooperation and coamplificationof developmental transcription factor genes

    in lung cancer. Proc Natl Acad Sci U S A2007;104:16663-8.

    Tang X, Sun M, Behrens C, et al.94.TITF-1gene amplification and protein ex-pression pattern identify adenocarcinomaof lung with worse prognosis. Presentedat the American Association for CancerResearch Annual Meet ing, San Diego, CA,April 1216, 2008. abstract.

    Merrick DT, Haney J, Petrunich S, et95.al. Overexpression of vascular endothelialgrowth factor and its receptors in bron-chial dypslasia demonstrated by quantita-tive RT-PCR analysis. Lung Cancer 2005;48:31-45.

    Keith RL, Miller YE, Gemmill RM, et96.al. Angiogenic squamous dysplasia inbronchi of individuals at high risk forlung cancer. Clin Cancer Res 2000;6:1616-25.

    Stricter RM. Out of the shadows: CXC97.chemokines in promoting aberrant lungangiogenesis. Cancer Prev Pres 2008 (inpress).

    Dowlati A, Gray R, Sandler AB, Schil-98.

    ler JH, Johnson DH. Cell adhesion mole-cules, vascular endothelial growth factor,and basic fibroblast growth factor in pa-tients with non-small cell lung cancertreated with chemotherapy with or with-out bevacizumab an Eastern Coopera-tive Oncology Group study. Clin CancerRes 2008;14:1407-12.

    Heymach JV, Hanrahan EO, Mann H,99.et al. Baseline VEGF as a potential predic-tive biomarker of vandetanib clinical bene-fit in patients with advanced NSCLC. J ClinOncol 2008;26:Suppl:426s. abstract .

    Giatromanolaki A, Koukourakis MI,100.Sivridis E, et al. Relation of hypoxia in-ducible factor 1 alpha and 2 a lpha in oper-

    able non-small cell lung cancer to angio-genic/molecular profile of tumours andsurvival. Br J Cancer 2001;85:881-90.

    Hoogsteen IJ, Marres HA, van der101.Kogel AJ, Kaanders JH. The hypoxic tu-mour microenvironment, patient selectionand hypoxia-modifying treatments. ClinOncol (R Coll Radiol) 2007;19:385-96.

    Pore N, Jiang Z, Gupta A, Cerniglia102.G, Kao GD, Maity A. EGFR tyrosine ki-nase inhibitors decrease VEGF expressionby both hypoxia-inducible factor (HIF)-1-independent and HIF-1-dependent mech-anisms. Cancer Res 2006;66:3197-204.

    Sandler A, Gray R, Perry MC, et al.103.Paclitaxelcarboplatin alone or with beva-cizumab for nonsmall-cell lung cancer.N Engl J Med 2006;355:2542-50. [Erratum,N Engl J Med 2007;356:318.]

    Viloria-Petit A, Crombet T, Jothy S,104.et al. Acquired resistance to the antitumoreffect of epidermal growth factor recep-tor-blocking antibodies in v ivo: a role foraltered tumor angiogenesis. Cancer Res2001;61:5090-101.

    Herbst RS, ONeill VJ, Fehrenbacher105.L, et al. Phase II study of eff icacy and safe-ty of bevacizumab in combination with

    chemotherapy or erlotinib compared withchemotherapy alone for treatment of re-current or refractory non small-cell lungcancer. J Clin Oncol 2007;25:4743-50.

    Heymach JV, Johnson BE, Prager D,106.et al. Randomized, placebo-controlledphase II study of vandetanib plus docetaxelin previously treated non small-cell lungcancer. J Clin Oncol 2007;25:4270-7. [Erra-

    tum, J Clin Oncol 2008;26:165-6.] Heymach J, Paz-Ares L, De Braud F,107.et al. Vandetanib alone or with paclitaxeland carboplatin as first-line treatment foradvanced NSCLC: a randomized phase IIstudy. J Clin Oncol (in press).

    Bhattacharjee A, Richards WG, Staun-108.ton J, et al. Classification of human lungcarcinomas by mRNA expression profilingreveals distinct adenocarcinoma subclass-es. Proc Natl Acad Sci U S A 2001;98:13790-5.

    Meyerson M, Carbone D. Genomic109.and proteomic profiling of lung cancers:lung cancer classification in the age oftargeted therapy. J Clin Oncol 2005;23:

    3219-26. Beer DG, Kardia SL, Huang CC, et al.110.

    Gene-expression profiles predict survivalof patients with lung adenocarcinoma.Nat Med 2002;8:816-24.

    Chen H-Y, Yu S-L, Chen C-H, et al.111.A five-gene signature and clinical outcomein nonsmall-cell lung cancer. N Engl JMed 2007;356:11-20.

    Potti A, Mukherjee S, Petersen R, et112.al. A genomic strategy to refine prognosisin early-stage nonsmall-cell lung can-cer. N Engl J Med 2006;355:570-80. [Er-ratum, N Engl J Med 2007;356:201-2.]

    Potti A, Dressman HK, Bild A, et al.113.Genomic signatures to guide the use of

    chemotherapeut ics. Nat Med 2006;12:1294-300. [Erratum, Nat Med 2007;13:1388.]

    Beane J, Sebastiani P, Whitf ield TH,114.et al. A prediction model for lung cancerdiagnosis that integrates genomic andclinical features. Cancer Prev Res 2008;1:56-64.

    Shedden K, Taylor JM, Enkemann115.SA, et al. Gene expression-based survivalprediction in lung adenocarcinoma: a multi-site, blinded validation study. Nat Med2008;14:822-7.

    Yu SL, Chen HY, Chang GC, et al.116.MicroRNA signature predicts survival andrelapse in lung cancer. Cancer Cell 2008;13:48-57.

    Weiss GJ, Bemis LT, Nakajima E, et117.al. EGFR regulation by microRNA in lungcancer: correlation with clinical responseand survival to gefitinib and EGFR ex-pression in cell lines. Ann Oncol 2008;19:1053-9.

    Gautschi O, Bigosch C, Huegli B, et118.al. Circulating deoxyribonucleic acid asprognostic marker in non-small-cell lungcancer patients undergoing chemotherapy.J Clin Oncol 2004;22:4157-64.

    Ramirez JL, Rosell R, Taron M, et al.119.

    The New England Journal of Medicine

    Downloaded from nejm.org on November 18, 2014. For personal use only. No other uses without permission.

    Copyright 2008 Massachusetts Medical Society. All rights reserved.

  • 8/10/2019 Molecular Origin Lung Cancer

    14/14

    n engl j med 359;13 www.nejm.org september 25, 20081380

    Molecular Origins of Cancer

    14-3-3sigma Methylation in pretreatmentserum circulating DNA of cisplatin-plus-gemcitabine-treated advanced non-small-cell lung cancer patients predicts survival.J Clin Oncol 2005;23:9105-12.

    Chen XQ, Stroun M, Magnenat JL,120.et al. Microsatellite alterations in plasmaDNA of small cell lung cancer patients.Nat Med 1996;2:1033-5.

    Belinsky SA, Grimes MJ, Casas E, et121.al. Predicting gene promoter methylationin non-small-cell lung cancer by evaluat-ing sputum and serum. Br J Cancer 2007;96:1278-83.

    Ludovini V, Pistola L, Gregorc V, et al.122.Plasma DNA, microsatellite alterations,and p53 tumor mutations are associatedwith disease-free sur vival in radically re-sected non-small cell lung cancer pa-tients: a study of the Perugia multidisci-plinary team for thoracic oncology. J ThoracOncol 2008;3:365-73.

    Maheswaran S, Sequist LV, Nagrath123.S, et al. Detection of mutations in EGFRincirculat ing tumor lung-cancer cells. N Engl

    J Med 2008;359:366-77.Yildiz PB, Shyr Y, Rahman JS, et al.124.

    Diagnostic accuracy of MALDI mass spec-trometric analysis of unfractionated serumin lung cancer. J Thorac Oncol 2007;2:893-901.

    Guo A, Villn J, Kornhauser J, et al.125.Signaling networks assembled by onco-genic EGFR and c-Met. Proc Natl Acad SciU S A 2008;105:692-7.

    Byers L, Nanjundan M, Girard L, et126.

    al. Reverse-phase protein array (RPPA) pro-filing of response to taxanes and epider-mal growth factor receptor (EGFR) in-hibitors identifies an inverse correlationbetween markers of sensitivity to doce-taxel and erlotinib in non-small cell lungcancer lines. Presented at the AmericanAssociation for Cancer Research AnnualMeeting, San Diego, CA, April 1216, 2008.

    abstract. Yanagisawa K, Tomida S, Shimada Y,127.Yatabe Y, Mitsudomi T, Takahashi T. A 25-signal proteomic signature and outcomefor patients with resected non-small-celllung cancer. J Natl Cancer Inst 2007;99:858-67.

    Patz EF Jr, Campa MJ, Gottlin EB,128.Kusmartseva I, Guan XR, Herndon JE II.Panel of serum biomarkers for the diag-nosis of lung cancer. J Clin Oncol 2007;25:5578-83.

    Taguchi F, Solomon B, Gregorc V, et129.al. Mass spectrometry to classify non-small-cell lung cancer patients for clinicaloutcome after treatment with epidermal

    growth factor receptor tyrosine kinase in-hibitors: a multicohort cross-institut ionalstudy. J Natl Cancer Inst 2007;99:838-46.

    Esteller M. Epigenetics in cancer.130.N Engl J Med 2008;358:1148-59.

    Tsou JA, Galler JS, Siegmund KD, et131.al. Identification of a panel of sensitiveand specific DNA methylation markersfor lung adenocarcinoma. Mol Cancer2007;6:70.

    Heist RS, Zhai R, Liu G, et a l. VEGF132.

    polymorphisms and survival in early-stage non-small-cell lung cancer. J ClinOncol 2008;26:856-62.

    Gregorc V, Hidalgo M, Spreafico A,133.et al. Germline polymorphisms in EGFRand survival in patients with lung cancerreceiving gefitinib. Clin Pharmacol Ther2008;83:477-84.

    Liu G, Gurubhagavatula S, Zhou W,134.

    et al. Epidermal growth factor receptorpolymorphisms and clinical outcomes innon-small-cell lung cancer pat ients treatedwith gefitinib. Pharmacogenomics J 2008;8:129-38.

    Simon G, Sharma A, Li X, et al. Fea-135.sibility and efficacy of molecular analysis-directed individualized therapy in ad-vanced non-small-cell lung cancer. J ClinOncol 2007;25:2741-6.

    Cobo M, Isla D, Massuti B, et a l. Cus-136.tomizing cisplatin based on quantitativeexcision repair cross-complementing 1mRNA expression: a phase III trial innon-small-cell lung cancer. J Clin Oncol2007;25:2747-54.

    Zhou X, Liu S, Kim ES, Herbst RS,137.Lee JJ. Bayesian adaptive design for tar-geted therapy development in lung cancer a step toward personalized medicine.Clin Trials 2008;5:181-93.

    Herbst RS, Lippman SM. Molecular138.signatures of lung cancer toward per-sonalized therapy. N Engl J Med 2007;356:76-8.Copyright 2008 Massachusetts Medical Society.

    FULLTEXTOFALLJOURNALARTICLESONTHEWORLDWIDEWEB

    Access to the complete text of theJournalon the Internet is free to all subscribers. To use this Web site, subscribers should goto theJournals home page (www.nejm.org) and register by entering their names and subscriber numbers as they appear ontheir mailing labels. After this one-time registration, subscribers can use their passwords to log on for electronic access to theentireJournalfrom any computer that is connected to the Internet. Features include a library of all issues since January 1993and abstracts since January 1975, a full-text search capacity, and a personal archive for saving articles and search results of

    interest. All articles can be printed in a format that is virtually identical to that of the typeset pages. Beginning 6 months afterpublication, the full text of all Original Articles and Special Articles is available free to nonsubscribers.

    The New England Journal of Medicine