ca paru1

8
Lung Cancer Biomarkers Present Status and Future Developments Philip T. Cagle, MD; Timothy Craig Allen, MD, JD; Randall J. Olsen, MD, PhD The publication of the ‘‘Molecular Testing Guideline for Selection of Lung Cancer Patients for EGFR and ALK Tyrosine Kinase Inhibitors: Guideline From the College of American Pathologists, International Association for the Study of Lung Cancer, and Association for Molecular Pathology’’ has now provided a guideline for biomarker testing for first-generation lung cancer tyrosine kinase inhibitors. Biomarker testing has forever altered the role of pathologists in the management of patients with lung cancer. Current, unresolved issues in the precision medicine of lung cancer will be addressed by the development of new biomarker tests, new drugs, and new test technologies and by improvement in the cost to benefit ratio of biomarker testing. (Arch Pathol Lab Med. 2013;137:1191–1198; doi: 10.5858/arpa.2013-0319-CR) I n April 2013, the College of American Pathologists (CAP), the International Association for the Study of Lung Cancer (IASLC), and the Association for Molecular Pathol- ogy (AMP) officially released the ‘‘Molecular Testing Guideline for Selection of Lung Cancer Patients for EGFR [epidermal growth factor receptor] and ALK [anaplastic lymphoma kinase] Tyrosine Kinase Inhibitors [TKIs].’’ 1 The CAP/IASLC/AMP guideline provides the first standardized evidence-based approach for performing biomarker testing to select patients with lung cancer for EGFR or ALK TKI therapy that is multidisciplinary and multicontinental in scope. Subsequent to the guideline release, the CAP is also releasing a template protocol for the reporting of lung cancer–predictive biomarkers as an addendum to anatomic pathology reports, and the guidelines, themselves, will be regularly updated. The number of predictive biomarkers to be tested for lung cancer, the number of targeted therapies, and the types of test methodologies are all expected to increase during upcoming months and years. The need for predictive biomarker testing on pathology specimens has forever altered the role of pathologists in the care of patients with lung cancer. This review provides an overview of current developments, expected developments, and related issues for lung cancer–predictive biomarker testing. HISTORICAL PERSPECTIVE For many years, lung cancer has been the leading cause of cancer deaths in the United States 2 and worldwide. 3 Most patients with lung cancer present with advanced disease, and conventional treatment options have been limited, resulting in an overall 5-year survival rate of only 10% to 15%, for many decades. 4,5 Approximately 85% of lung cancers are non–small cell lung cancers (NSCLCs), traditionally divided into 3 major cell types: adenocarcinoma (approximately 50%), squa- mous cell carcinoma (approximately 35%), and large cell carcinoma (approximately 15%, although this is a dimin- ishing cell type category). 6,7 About 70% of NSCLCs present with advanced disease not considered curable by surgical resection, either locally advanced (stage IIIB) or often with metastatic disease (stage IV). Clinical stage IIIB NSCLCs are associated with a 5-year survival rate of 7% and stage IV NSCLCs, with a 5-year survival rate of 2%. 8 Conven- tional therapy for stage IV NSCLC is doublet chemother- apy that includes cisplatin or carboplatin. Affected patients may additionally receive radiation therapy. Of the patients with lung cancer who initially respond to first-line therapy, nearly all subsequently experience disease progression. These patients may receive second-line therapy, or possibly more lines of therapy, in an attempt to control their disease. Eventually, virtually all of these patients die from lung cancer as reflected in the statistics mentioned previously. 9–16 Given the dismal prognosis and limited treatment options for patients with advanced-stage lung cancer, it is not surprising that the US Food and Drug Administration (FDA) approval of targeted molecular therapies for advanced-stage lung cancer has been a game changer for these patients. The first generation of EGFR TKIs approved for treatment of advanced-stage lung cancer includes gefitinib (Iressa; AstraZeneca, London, United Kingdom) and erlotinib (Tarceva; Genentech, South San Francisco, California, and OSI Pharmaceuticals, Long Island, New York). Already Accepted for publication May 28, 2013. From the Department of Pathology and Genomic Medicine, The Methodist Hospital, Houston, Texas, and the Department of Pathology and Laboratory Medicine, Weill Medical College of Cornell University, New York, New York (Drs Cagle and Olsen); and the Department of Pathology, The University of Texas Health Science Center at Tyler (Dr Allen). The authors have no relevant financial interest in the products or companies described in this article. Presented at the New Frontiers in Pathology: An Update for Practicing Pathologists meeting; Homestead Resort; August 3–5, 2012; Glen Arbor, Michigan. Reprints: Philip T. Cagle, MD, Department of Pathology and Genomic Medicine, The Methodist Hospital, 6565 Fannin St, Main Bldg, Room 227, Houston, Texas 77030 (e-mail: [email protected]). Arch Pathol Lab Med—Vol 137, September 2013 Lung Cancer Biomarkers: Present and Future—Cagle et al 1191

Upload: maya-rustam

Post on 03-Dec-2015

1 views

Category:

Documents


0 download

DESCRIPTION

Ca paru

TRANSCRIPT

Page 1: Ca Paru1

Lung Cancer Biomarkers

Present Status and Future Developments

Philip T. Cagle, MD; Timothy Craig Allen, MD, JD; Randall J. Olsen, MD, PhD

� The publication of the ‘‘Molecular Testing Guideline forSelection of Lung Cancer Patients for EGFR and ALKTyrosine Kinase Inhibitors: Guideline From the College ofAmerican Pathologists, International Association for theStudy of Lung Cancer, and Association for MolecularPathology’’ has now provided a guideline for biomarkertesting for first-generation lung cancer tyrosine kinaseinhibitors. Biomarker testing has forever altered the role ofpathologists in the management of patients with lungcancer. Current, unresolved issues in the precisionmedicine of lung cancer will be addressed by thedevelopment of new biomarker tests, new drugs, andnew test technologies and by improvement in the cost tobenefit ratio of biomarker testing.

(Arch Pathol Lab Med. 2013;137:1191–1198; doi:10.5858/arpa.2013-0319-CR)

In April 2013, the College of American Pathologists (CAP),the International Association for the Study of Lung

Cancer (IASLC), and the Association for Molecular Pathol-ogy (AMP) officially released the ‘‘Molecular TestingGuideline for Selection of Lung Cancer Patients for EGFR[epidermal growth factor receptor] and ALK [anaplasticlymphoma kinase] Tyrosine Kinase Inhibitors [TKIs].’’ 1 TheCAP/IASLC/AMP guideline provides the first standardizedevidence-based approach for performing biomarker testingto select patients with lung cancer for EGFR or ALK TKItherapy that is multidisciplinary and multicontinental inscope. Subsequent to the guideline release, the CAP is alsoreleasing a template protocol for the reporting of lungcancer–predictive biomarkers as an addendum to anatomicpathology reports, and the guidelines, themselves, will be

regularly updated. The number of predictive biomarkers tobe tested for lung cancer, the number of targeted therapies,and the types of test methodologies are all expected toincrease during upcoming months and years. The need forpredictive biomarker testing on pathology specimens hasforever altered the role of pathologists in the care of patientswith lung cancer. This review provides an overview ofcurrent developments, expected developments, and relatedissues for lung cancer–predictive biomarker testing.

HISTORICAL PERSPECTIVE

For many years, lung cancer has been the leading cause ofcancer deaths in the United States2 and worldwide.3 Mostpatients with lung cancer present with advanced disease,and conventional treatment options have been limited,resulting in an overall 5-year survival rate of only 10% to15%, for many decades.4,5

Approximately 85% of lung cancers are non–small celllung cancers (NSCLCs), traditionally divided into 3 majorcell types: adenocarcinoma (approximately 50%), squa-mous cell carcinoma (approximately 35%), and large cellcarcinoma (approximately 15%, although this is a dimin-ishing cell type category).6,7 About 70% of NSCLCs presentwith advanced disease not considered curable by surgicalresection, either locally advanced (stage IIIB) or often withmetastatic disease (stage IV). Clinical stage IIIB NSCLCsare associated with a 5-year survival rate of 7% and stageIV NSCLCs, with a 5-year survival rate of 2%.8 Conven-tional therapy for stage IV NSCLC is doublet chemother-apy that includes cisplatin or carboplatin. Affected patientsmay additionally receive radiation therapy. Of the patientswith lung cancer who initially respond to first-line therapy,nearly all subsequently experience disease progression.These patients may receive second-line therapy, or possiblymore lines of therapy, in an attempt to control theirdisease. Eventually, virtually all of these patients die fromlung cancer as reflected in the statistics mentionedpreviously.9–16

Given the dismal prognosis and limited treatment optionsfor patients with advanced-stage lung cancer, it is notsurprising that the US Food and Drug Administration (FDA)approval of targeted molecular therapies for advanced-stagelung cancer has been a game changer for these patients.

The first generation of EGFR TKIs approved for treatmentof advanced-stage lung cancer includes gefitinib (Iressa;AstraZeneca, London, United Kingdom) and erlotinib(Tarceva; Genentech, South San Francisco, California, andOSI Pharmaceuticals, Long Island, New York). Already

Accepted for publication May 28, 2013.From the Department of Pathology and Genomic Medicine, The

Methodist Hospital, Houston, Texas, and the Department ofPathology and Laboratory Medicine, Weill Medical College ofCornell University, New York, New York (Drs Cagle and Olsen);and the Department of Pathology, The University of Texas HealthScience Center at Tyler (Dr Allen).

The authors have no relevant financial interest in the products orcompanies described in this article.

Presented at the New Frontiers in Pathology: An Update forPracticing Pathologists meeting; Homestead Resort; August 3–5,2012; Glen Arbor, Michigan.

Reprints: Philip T. Cagle, MD, Department of Pathology andGenomic Medicine, The Methodist Hospital, 6565 Fannin St, MainBldg, Room 227, Houston, Texas 77030 (e-mail: [email protected]).

Arch Pathol Lab Med—Vol 137, September 2013 Lung Cancer Biomarkers: Present and Future—Cagle et al 1191

Page 2: Ca Paru1

since the 1980s it was known that most NSCLCsoverexpress EGFR,17 but early clinical trials of EGFR TKIsaffected only a small percentage of patients with lungcancer. In 2004, several separate investigators reported thatthe presence of somatic mutations of EGFR gene in a lungcancer predicted the likelihood of its response to EGFRTKIs.18–20 Beginning in 2009, a number of clinical trials havereported better response rates and progression-free survivalfor patients with advanced-stage NSCLC with EGFRmutations who received EGFR TKIs compared to conven-tional therapy.21–23 These clinical trials were conducted withpatients with advanced lung cancer who had significantimprovement in progression-free survival, but no demon-strated improvement in overall survival, possibly because ofthe crossover design of these studies in which many of thepatients were first treated with chemotherapy.1

Subsequent studies have demonstrated that almost all ofthe EGFR mutations are in adenocarcinomas or NSCLCswith an adenocarcinoma component, including adenosqua-mous carcinoma and solid subtypes of adenocarcinomainitially misinterpreted histologically as squamous cellcarcinomas but proven to be adenocarcinoma after furtherworkup.1,24,25 Clinical evidence also demonstrated that EGFRmutation analysis was the most reliable method ofdetermining which NSCLCs have EGFR mutations, and ofpredicting response to first-generation TKI therapy, asopposed to EGFR copy number detection by fluorescencein situ hybridization (FISH) or protein expression byimmunohistochemistry (IHC).1,4,5,25–27

In 2007, translocation of the anaplastic lymphoma kinasegene (ALK) with echinoderm microtubule-associated pro-tein-like 4 gene (EML4) was first reported in pulmonaryadenocarcinomas.28 Later studies29 showed that there weremany variants of the EML4-ALK rearrangement in additionto ALK fusion with other partners. When clinical trialsdemonstrated that crizotinib therapy improved responserate and progression-free survival for patients with ad-vanced NSCLC with ALK rearrangements, the FDA grantedaccelerated approval of the ALK TKI crizotinib (Xalkori;Pfizer, New York, New York).30,31 Concurrently, the FDAapproved a specific companion test (Vysis ALK Break-ApartFISH Probe Kit, Abbott Molecular, Des Plaines, Illinois) thathad been used to select patients for therapy with Xalkori inthe clinical trials.32 To date, cytogenetic techniques such asFISH have been generally considered a better method fordetecting chromosomal rearrangements, such as thoseinvolving ALK. It has been observed that reverse transcrip-tion–polymerase chain reaction or other methods thatrequire primers for each possible fusion variant may havefalse negatives.31,33

THE CAP/IASLC/AMP GUIDELINE ‘‘MOLECULARTESTING GUIDELINE FOR SELECTION OF LUNG

CANCER PATIENTS FOR EGFR AND ALK TYROSINEKINASE INHIBITORS’’

In the past several years, it became obvious that a subsetof patients with lung cancer have responded to the first-generation TKI therapies. However, there were no uniformguidelines for performing biomarker testing to selectpatients for treatment that was based on an in-depth reviewand grading of evidence and that incorporated input andvetting across the relevant medical disciplines on aworldwide basis. Therefore, the CAP, IASLC, and AMPset out to create standardized international guidelines for

lung cancer biomarker testing with input from multipleother stakeholders in an extended and exhaustive process asdescribed in the opening sections of the guidelines.1 Thiswork was conducted under the auspices of the CAPPathology and Laboratory Quality Center.1

The CAP/IASLC/AMP guideline focuses on the lungcancer–predictive biomarkers corresponding to the first-generation EGFR TKIs (gefitinib and erlotinib) and ALK TKI(crizotinib), which are clinically validated and FDA approvedfor treatment of advanced-stage lung cancers.1 Per theguidelines, EGFR and ALK molecular testing of advanced-stage lung cancers is recommended to select patients forEGFR or ALK TKI therapy and, after the diagnosis ofprimary lung adenocarcinoma is made (with the caveatmentioned below), tissue should be prioritized for biomark-er testing. For limited specimens, EGFR testing should beprioritized first and ALK testing should be prioritizedsecond over other molecular markers. Clinical criteria suchas sex, ethnicity, and smoking status are not recommendedfor selecting patients for lung cancer–predictive biomarkertesting, but adenocarcinoma cell type is a basis for selectingfor biomarker testing. With small biopsy or cytologyspecimens, individual judgment is permissible and bio-marker testing may be warranted in situations in which adiagnosis of adenocarcinoma cannot be excluded.1

A wide range of sample types, including cytologyspecimens, and fixatives (formalin-fixed paraffin-embedded,fresh, frozen, and alcohol) are allowable for biomarkertesting. Mutation analysis, using a validated method withsufficient performance characteristics, is recommended forEGFR mutation testing. KRAS mutation testing is notrecommended as a sole determinant of EGFR TKI therapybecause most lung cancers that lack KRAS mutations alsolack EGFR mutations. FISH assay using dual-labeled break-apart probes is recommended for ALK translocation testing.Carefully validated ALK IHC can be used as a screeningmethod to select specimens for ALK FISH testing. Thereader is referred to the published guideline for additionaldetails and the complete recommendations, suggestions,and expert consensus opinions.1

UNRESOLVED ISSUES, CAVEATS,AND FUTURE OBJECTIVES

The results of clinical trials with first-generation TKIs andthe release of the CAP/IASLC/AMP ‘‘Molecular TestingGuideline for Selection of Lung Cancer Patients for EGFRand ALK Tyrosine Kinase Inhibitors’’ have offered muchpromise and generated considerable excitement amongpatients, their families, their physicians, and the media.1

Looking forward, there are multiple unresolved issues,caveats, and future objectives that must be addressed:

1. The original clinical trials of first-generation EGFR andALK TKIs were performed with patients with advanced-stage lung cancer and, therefore, the approvals are forpatients with stage IIIB and IV lung cancer. Since manypatients with earlier-stage lung cancer later have lungcancer recurrence, and many ultimately die from theirdisease, this raises the question of whether or notpredictive biomarker testing should be done in theseearlier-stage lung cancers, either to have the data fortreatment when recurrence occurs or to use as anadjuvant therapy during initial treatment.1

2. EGFR mutations are identified in only approximately15% of NSCLCs in white persons in the United States

1192 Arch Pathol Lab Med—Vol 137, September 2013 Lung Cancer Biomarkers: Present and Future—Cagle et al

Page 3: Ca Paru1

and in 20% of NSCLCs in African American persons.ALK rearrangements are found in only approximately 4%of adenocarcinomas in white persons in the UnitedStates.1 Therefore, targeted therapies and correspondingbiomarkers are still needed for most pulmonary adeno-carcinomas.27

3. Virtually all NSCLCs develop acquired resistance to first-generation EGFR TKIs or crizotinib after a period ofinitial response that may last for months. This acquiredor secondary resistance results from secondary mutationsin the EGFR or ALK gene or to a variety of othermechanisms.1,25 Second-line or third-line therapies forthese patients might include second-generation TKIs ordrugs directed at other actionable targets.34,35

4. EGFR mutations and ALK rearrangements are found inpulmonary adenocarcinomas, including adenosquamouscarcinomas and other variants, but are found uncom-monly, if at all, in pure squamous cell carcinomas or puresmall cell carcinomas.4,5,24,26,27,36 Therefore, targetedtherapies and corresponding biomarkers are needed forlung cancer cell types other than adenocarcinoma.25

5. Investigations into targeted therapy of lung cancer stemcells may provide a novel alternative to treating thesepatients.37

6. New technologies are altering the paradigm for predic-tive biomarker testing.1,25

7. Reimbursement for predictive biomarker testing is animportant issue if testing is going to be done and,especially, if it is going to be done on a reflex basis.1

THERAPY IN EARLY-STAGE LUNG CANCER

Only approximately 30% of NSCLCs are diagnosed in anearly stage (stage I, II, or IIIA) with limited disease. Mostaffected patients are treated with surgical resection and mayalso receive adjuvant therapy based on various proto-cols.16,38,39 However, despite treatment during early stage, alarge percentage of these patients will nevertheless haverelapse, with disease progressing to an advanced stage, andthey will eventually die from their lung cancer (5-year survivalrate by clinical stage is 50% for stage IA, 43% for stage IB,36% for stage IIA, 25% for stage IIB, and 19% for stage IIIA).8

Since most patients with early-stage lung cancer willeventually have relapse with disease progression, the CAP/IASLC/AMP guideline encourages EGFR and ALK testing oflung cancers at the time of diagnosis for patients presentingwith stage I, II, or III disease.1 Alternatively, if testing is notperformed in these early-stage cancers, the guidelinesencourage the retaining of cancer tissue for future biomarkertesting should the patient’s condition progress to anadvanced stage.1

Biomarker testing of the lung cancer tissue may provide abasis for TKI therapy when the patient’s conditionprogresses to an advanced stage. It may also form the basisfor TKI therapy as an adjuvant therapy at the time of initialdiagnosis and treatment in the early stage.1 Severalstudies40–42 are investigating the use of first-generationEGFR TKIs as adjuvant therapy in early-stage lung cancers,including the RADIANT and SELECT trials.

MOLECULAR TARGETED THERAPIESUNDER INVESTIGATION

Cetuximab (Erbitux; Bristol-Myers Squibb, New York,New York, and Eli Lilly and Company, Indianapolis,Indiana) has been approved by the FDA for treatment of

advanced colon adenocarcinoma and advanced head andneck squamous cell carcinoma.43–47 In 2011 and 2012,subgroup analysis of the First-Line Erbitux in Lung Cancer(FLEX) phase III clinical trial48–50 found that high expressionwith EGFR IHC (score of 200 or more) using the DakopharmDx Kit (Glostrup, Denmark) correlated with increasedoverall survival for patients with advanced NSCLC whowere receiving first-line platinum-based chemotherapy pluscetuximab, compared to chemotherapy alone, for lungsquamous cell carcinomas and adenocarcinomas. Thescoring system for EGFR IHC in this context has since beenvalidated.51 Cetuximab may offer a new therapy for bothlung adenocarcinomas and lung squamous cell carcinomas,based on a different predictive biomarker test, EGFR IHC asopposed to EGFR mutation testing.52

Patients receiving first-generation EGFR TKIs eventuallydevelop acquired resistance to their drug. Second-genera-tion EGFR TKIs are under investigation as additional lines oftherapy when acquired resistance develops or as apotentially more effective first-line therapy. These drugsare ERBB family blockers and, compared to first-generationEGFR TKIs, they typically exhibit higher affinity for thetarget, irreversible binding, and inhibition of more than 1target in the ERBB family of receptors. In addition, some ofthe second-generation drugs may also bind to EGFRreceptors with mutations of acquired resistance to thefirst-generation EGFR TKIs.53–56

Three of these ERBB family blockers under investigationare afatinib (BIBW2992; Boehringer Ingelheim, Ingelheim,Germany),54,56 dacomitinib,57 and XL647,58 Afatinib irre-versibly binds to HER2/neu, HER4, and EGFR, even whenthe most common acquired resistance mutation (T790M) ispresent. Dacomitinib is also a pan-ERBB family blocker thatcan potentially achieve response in tumors harboring theT790M mutation.

Some TKIs inhibit more than 1 kinase. Crizotinib,currently used for ALKþ adenocarcinomas, is a primeexample. In addition to ALK, crizotinib inhibits ROS1,MET, and RON.59,60 ROS1 translocations occur in 1% to 2%of NSCLCs, with several different fusion partners reported.Approval of crizotinib for treatment of ROS1þ adenocarci-nomas is generally expected in the not too distant future.60,61

There are multiple other potentially druggable targetswith corresponding predictive biomarkers in lung adeno-carcinomas, including in the signaling pathways down-stream of the ERRB receptors. Other drugs undergoingclinical trials include (1) the mammalian target of rapamycin(mTOR) inhibitor everolimus, the phosphoinositide-3 ki-nase (PI3K) and mTOR inhibitor BEZ235, the PI3Kinhibitors GDC-0941 and XL147, and the AKT inhibitorMK-220662–65; (2) inhibitors and antibodies for c-MET andits ligand, hepatocyte growth factor66; tivantinib as second-line therapy in patients with advanced nonsquamousNSCLC in the MARQUEE (Met Inhibitor ARQ 197 plusErlotinib versus Erlotinib plus placebo in NSCLC) trial67,68;the JAK (Janus kinase) inhibitors enzastaurin and AZD1480and the STAT inhibitor NSC-74338069–71; inhibitors of MEK(mitogen-activated protein kinase kinase),72–74 vandetanibfor RET (rearranged during transfection),75,76 and dasatinibfor Src.77,78

KRAS is the most frequently mutated oncogene in lungadenocarcinomas, occurring in about 30% of cases, mostlyin smokers. There are currently no direct inhibitors of KRAS,although there are inhibitors of targets downstream toKRAS.79

Arch Pathol Lab Med—Vol 137, September 2013 Lung Cancer Biomarkers: Present and Future—Cagle et al 1193

Page 4: Ca Paru1

Cetuximab in combination with first-line platinum-basedchemotherapy has been reported to improve overall survivalfor patients with lung squamous cell carcinomas that havehigh total EGFR expression by IHC (score of 200 or more)using the Dako pharmDx kit, compared to chemotherapyalone.48–52

Potential targets for squamous cell carcinoma underinvestigation include members of the PI3K pathway, thefibroblast growth factor receptor, and the discoidin domainreceptor.80–83 Potential drugs under investigation for smallcell lung cancer include the mTOR inhibitors everolimusand temsirolimus.84–86

MOLECULAR TARGETINGOF LUNG CANCER STEM CELLS

Stem cells in lung cancer have been postulated for morethan 3 decades,87 and in the last decade they have beenincreasingly examined for the presence of possible diagnos-tic and therapeutic benefits. Lung cancer stem cells havespecific characteristics of all stem cells, including chemore-sistance and radioresistance, the ability to self-renew, theability to produce a large number of multilineage progeny,and slow proliferation.88,89 Postchemoradiotherapy expres-sion of lung cancer stem cell markers correlates with poorprognosis in patients with lung cancer.88 Research iscontinuing to clarity the critical role lung cancer stem cellsplay in metastases, drug resistance, and tumor regenera-tion.37 The predominant use of mouse models, as well aslung cancer stem cells’ slow cycling and asymmetric celldivision, make their identification difficult; and studies ofpotential treatments targeting lung cancer stem cells are allthe more challenging.37 Identification of molecular targetsfor effective lung cancer stem cell therapy requires thedifferentiation of lung cancer stem cells from normalmultipotent stem cells. This differentiation might beaccomplished by using magnetic bead isolation or flowcytometry to identify unique cell type markers.90

Treatment regimens for cancers have traditionally as-sumed that all cancer cells have equal malignant potential,so the concept that cancers are driven by cancer stem cellshas significant clinical implications.91 Researchers are nowactively seeking therapies that target stem cells, and cancerstem cell inhibitory mechanisms, such as blocking stem cellfactor, antagonists of ABCG2 pumping activity, and Notchinhibitors, are becoming increasingly studied.91 Hedgehog,Wng, and Notch pathways have been found to be critical tostem cell regulation, and might provide appropriate therapytargets.37 The Hedgehog signaling pathway is essential fordetermining whether a cell undergoes self-renewal ordifferentiation.92 Wnt signaling is necessary for embryogen-esis and homeostatic maintenance of adult tissues, and itsactivation has been shown to significantly enhance lungcancer proliferation, migration, colony formation, and drugresistance.37,92 Approximately one-third of lung cancershave an activated Notch pathway, and its presence has beenshown to indicate a significantly worse prognosis in patientswho concomitantly lack TP53 mutations.37 Studies of lungcancer stem cell–targeted therapy based on these pathwaysis ongoing.92 Ongoing studies of potential lung cancer stemcell–targeted therapies are also ongoing. For example, astudy showing significantly increased Rac1 guanosinetriphosphatase activity in lung cancer cells that haveundergone epithelial-mesenchymal transition suggests thattargeting Rac1 might provide a more effective lung cancer

therapy by eliminating cancer stem cell subpopulations andby blocking non–cancer stem cell to cancer stem celltransition and eliminating cancer stem cell subpopula-tions.93 It has also been suggested that ALDH1A1þ lungcancer stem cells may cause EGFR TKI resistance.94 Also,another study95 suggests that an existing phenothiazine-likeantipsychotic drug, trifluoperazine, has the ability to down-regulate cancer stem cell markers CD44 and CD133 and assuch might possess anti–lung cancer stem cell properties.Finally, the concept of a single lung cancer stem cell hasbeen complicated by the possibility of an associatedmesenchymal stem cell that might play a role in lungcancer tumorigenesis and progression by producing ormaintaining a cancer-permissive microenvironment.96

ADVANCES IN NEXT-GENERATION TECHNOLOGY

Before the debut of benchtop next-generation sequencinginstruments, clinical laboratories had limited options foridentifying the presence or absence of gene mutations intumors. Many molecular pathologists developed tests usingtraditional technologies, such as pyrosequencing, Sangerdideoxynucleotide chain termination sequencing, or real-time polymerase chain reaction.97 Although these assaysgenerate the results needed for patient care, they are limitedby a single-target-gene approach.98 That is, a separateassay—and in some cases, such as EGFR, multiple assays—must be performed for each biomarker. As outlined above,testing for multiple biomarkers is already considered to bethe standard of care for lung cancer, and additional tests willbe needed as new predictive biomarkers and targetedtherapies are discovered. Thus, when specimen volumesare limited, such as commonly occurs with small biopsies orcytopathology procedures, there may be insufficient diag-nostic material available to perform all molecular diagnos-tics testing.6,99 For this reason, many laboratories are nowdeveloping strategies to optimally triage tissue for histopa-thology examination, immunohistochemistry, FISH, andmolecular testing with direction provided by the CAP/IASLC/AMP guidelines.98 In this context, next-generationsequencing technologies are significantly advancing molec-ular diagnostics by enabling the design of highly multi-plexed assays.100,101

Several next-generation sequencing platforms are com-mercially available. The technologies vary in the type ofDNA molecule used as a sequencing template, thechemistry underlying the sequencing reaction, and theamount of sequence data generated per run. The instru-ments most commonly used in clinical laboratories are the454 GS Junior (454 Life Sciences, Roche, Branford,Connecticut), Ion Torrent Personal Genome Machine(Ion Torrent Systems, Life Technologies, San Francisco,California), and MiSeq Personal Sequencer (Illumina Inc,San Diego, California). The 454 was first to enter themarket. It uses emulsion polymerase chain reaction toclonally amplify DNA fragments that are sequenced viasequencing-by-synthesis technology.102 The Ion Torrent isbased on a similar technology, but rather than usingfluorescence to determine the nucleotide sequence, itdetects Hþ ions that are released during base incorpora-tion.103 In comparison, the MiSeq uses bridge amplificationto clonally amplify DNA fragments before sequencing.103

The MassARRAY Analyzer (Sequenom Inc, San Diego,California) is another innovative technology. It uses massspectrometry to determine the sequence of amplified gene

1194 Arch Pathol Lab Med—Vol 137, September 2013 Lung Cancer Biomarkers: Present and Future—Cagle et al

Page 5: Ca Paru1

fragments.104 Of note, investigators have now developednovel strategies to detect EML4-ALK gene rearrangementson the MassARRAY Analyzer, making it possible toconcurrently evaluate EGFR gene mutations and ALK generearrangements during a single run.105 A similar strategycould be designed to also detect ROS1 or other generearrangements. More recently, single-molecule sequenc-ing platforms, such as the PacBio RS (Pacific Biosciences,Menlo Park, California), have entered the researchenvironment as powerful tools for whole genome se-quencing.106 Although these instruments generate high-quality sequence data, each varies in key performancecharacteristics such as throughput, read length, error rate,hands-on time, run time, instrument cost, and reagentcost.107,108 We recommend that potential users carefullyevaluate their options, considering the intended clinicalapplication and individual laboratory needs. To assistpathologists in designing highly multiplexed gene muta-tion assays, reagent kits licensed for research use only arenow available. Also, an expert panel for next-generationsequencing recently suggested guidelines for test valida-tion, quality control, proficiency testing, and referencematerials,109 and the CAP has published a next-generationsequencing checklist (www.cap.org, June 21, 2013).

Although still in its infancy as a diagnostic tool, wholegenome sequencing has the potential to truly personalizepatient care. Importantly, the long-sought $1000 genomewill soon be a reality, and advances in automation andbioinformatics are making cancer genomics an increasinglytractable tool in the molecular diagnostics laboratory.110 Twoinvestigations80,111 recently reported the whole genomesequences of 178 lung squamous cell carcinomas and of183 lung adenocarcinomas. These incredibly rich data setsclearly reveal the genomic complexity of lung cancer. Forexample, the frequent presence of EGFR, KRAS, and BRAFmutations was confirmed in lung adenocarcinoma.111

Unexpectedly though, mutations were also identified inseveral other genes including U2AF1, RBM10, and ARID1A.In total, 25 genes were shown to have a statisticallysignificant number of mutations in lung adenocarcinoma.111

Each represents a possible target for new therapeutics. Incomparison, the squamous cell carcinoma investigationidentified only 11 genes with recurrent mutations.80

However, multiple signaling pathways were significantlyaltered. These include gene pathways implicated in oxidativestress response, apoptotic signaling, and squamous celldifferentiation.80 Of note, many of the alterations identifiedin squamous cell carcinoma of the lung are shared withthose occurring in squamous cell carcinoma of the head andneck, suggesting a possible common molecular biology.Taken together, these genomic investigations show thatpotential therapeutic targets can be found in nearly everylung tumor. Furthermore, researchers are expanding ourgenomic understanding of lung cancer to include epigenetic,transcriptomic, proteomic, and metabolomic signatures.112

When clinical phenotypes based on genomic informationcan be reliably predicted, then increasingly personalizedtherapeutic strategies will become possible.

Inasmuch as a molecular diagnostics laboratory can generatetremendous amounts of data by using next-generationsequencing, information management and interpretation is asignificant challenge. Given the magnitude of the humangenome (approximately 3 billion base pairs encoding approx-imately 20 000 genes), some investigative pathologists are nowbuilding bioinformatics and statistics support into their clinical

laboratories. However, before genomics can be performed as aroutine test, automated bioinformatics pipelines and analysisalgorithms must be developed. Currently, whole genomesequence data cannot be interpreted within a time frame that isoptimal for patient care. Furthermore, the functional conse-quence of many gene polymorphisms remains unknown.Molecular pathogenesis researchers must investigate allcommon gene alterations, defining which are driver mutationsand which are bystander mutations. Coordinated efforts togenerate a comprehensive reference human genome sequenceand cancer genome database (http://cancergenome.nih.gov,http://cancercommons.org, and http://icgc.org; each accessedMay 27, 2013) have begun filling these knowledge gaps.

ADVANCES IN BIOMARKERIMMUNOHISTOCHEMISTRY

Traditional EGFR IHC detects total EGFR proteinexpression, but does not differentiate between wild typeand mutations so it is not recommended as a basis forselecting patients for EGFR TKI therapy.98 Investigationshave examined antibodies to proteins associated with the 2primary mutations, which represent 90% of EGFR mutationsthat are associated with response to first-generation EGFRTKIs. These antibodies may be useful in screening for mostEGFR mutations but will not detect the less frequent EGFRmutations.113–115

As discussed previously, total EGFR protein expression byIHC may be the predictive biomarker used if cetuximab isapproved for lung cancer therapy. High expression withEGFR IHC (score of 200 or more) using the Dako pharmDxkit correlated with increased overall survival for patientswith lung cancer—specifically, adenocarcinoma and squa-mous cell carcinoma—receiving first-line platinum-basedchemotherapy plus cetuximab compared to chemotherapyalone.48–50 The EGFR expression scoring system has alreadybeen validated for this particular setting.51,52

Immunohistochemistry does not demonstrate ALK expres-sion in most tissues that lack an ALK fusion gene. When anALK fusion gene is present, IHC usually reveals increasedALK expressions. Well-known examples include lymphomaand inflammatory myofibroblastic tumor. Probably because ofthe relatively low expression of ALK protein in lung cancerswith ALK translocations, false-negative IHC results have beenobserved in lung cancers that were shown to have ALK fusiongenes by other methods.1,25,116 Several procedures have beenused to enhance the sensitivity of IHC for ALKþ lung cancers,but ALK antibodies that are highly specific and sensitive toALK expression in lung cancers have been developed. WhileALK expression is known to correlate with ALK transloca-tions with these antibodies, clinical trials validating ALK IHCas a predictive biomarker for patient outcomes with crizotinibtherapy have not been performed.1,25,116–120 The CAP/IASLC/AMP guidelines note that screening for ALK translocationswith the new sensitive ALK antibodies may potentially beused to identify lung cancers that should then have ALKtranslocations confirmed with FISH.1

PAYMENT FOR PREDICTIVE BIOMARKER TESTSAND THE FUTURE OF TARGETED THERAPY

A common and important, but as yet unanswered,question is how pathologists and laboratories will be paidfor performing lung cancer biomarker tests. Many oncolo-gists and patients are now requesting biomarker testing and,in some hospitals, reflex testing has been implemented as a

Arch Pathol Lab Med—Vol 137, September 2013 Lung Cancer Biomarkers: Present and Future—Cagle et al 1195

Page 6: Ca Paru1

policy. The number of requests is expected to increase andthe addition of new biomarkers for which testing may bedone is also expected to increase. For patients with lungcancer with a very bleak prognosis, for their families, and fortheir oncologists, it is difficult to put any price on potentialadditional months of life, particularly based on taking a pillwithout the significant inconveniences and side effects ofchemotherapy, even if ultimately there is relapse. Currently,pathologists and laboratories have the option of billingMedicare or private payers for each individual test that isordered separately, or of billing for testing as part of a reflextesting protocol at their institution. Some payers areconsidering bundled payments for biomarker testing withresulting treatment based on the average cost of care, forwhich providers accept an increased risk for overutiliza-tion.121

Whether, under what circumstances, and to what extentMedicare and private payers will pay for these tests,however, is currently unknown. No standard billing strategyexists for lung cancer biomarker test payment by Medicareor private payers. Cost-effectiveness analysis, evaluatingboth the clinical and economic effect of a test or treatment,is currently used by the United Kingdom’s National HealthService and, with looming health care reform, may soon beused in the United States to determine whether moleculartherapies and biomarkers are covered.122,123 If cost-effec-tiveness analyses become an accepted method in the UnitedStates for payers to determine coverage and price of lungcancer biomarker tests, evidence-based literature regardingthe value of these tests in patients with lung cancer will beneeded to influence payers’ payment determinations.However, advances in testing and treatment discussed,including less costly tests with improvements in turnaroundtime (eg, IHC or next-generation technology) and betteroutcomes from new therapies (new drugs and new targets)and treatment strategies (combined or sequential targets,adjuvant therapy in earlier stages), may significantly alterthe cost-benefit dynamic in the future.

CONCLUSION

The advent of predictive biomarker testing on tissuesamples for targeted therapy has forever altered the role ofthe pathologist in the management of patients with lungcancer. The role of the pathologist in biomarker testing,including very direct participation with IHC, has potential togrow. Patients with lung cancer and their families are nowmore aware of who pathologists are and the critical role thatthey have in their health care. New tests, new testtechnologies, and overall advances in precision medicinewill likely improve the cost to benefit ratio for biomarkertesting which, along with the powerful desire from patientsand physicians to improve survival and quality of life,suggest that predictive biomarker testing will expand as aroutine component of cancer care.

References

1. Lindeman NI, Cagle PT, Beasley MB, et al. Molecular testing guideline forselection of lung cancer patients for EGFR and ALK tyrosine kinase inhibitors:guideline from the College of American Pathologists, International Association forthe Study of Lung Cancer, and Association for Molecular Pathology [publishedonline ahead of print April 3, 2013]. Arch Pathol Lab Med. 2013. doi:10.5858/arpa.2012-0720-OA.

2. Siegel R, Naishadham D, Jemal A. Cancer statistics, 2012. CA Cancer JClin. 2012;62(1):10–29.

3. Youlden DR, Cramb SM, Baade PD. The international epidemiology of lungcancer: geographical distribution and secular trends. J Thorac Oncol. 2008;3(8):819–831.

4. Cagle PT, Allen TC, Dacic S, et al. Revolution in lung cancer: newchallenges for the surgical pathologist. Arch Pathol Lab Med. 2011;135(1):110–116.

5. Cagle PT, Dacic S. Lung cancer and the future of pathology. Arch PatholLab Med. 2011;135(3):293–295.

6. Travis WD, Brambilla E, Noguchi M, et al. Diagnosis of lung cancer insmall biopsies and cytology: implications of the 2011 International Associationfor the Study of Lung Cancer/American Thoracic Society/European RespiratorySociety Classification. Arch Pathol Lab Med. 2013;137(5):668–684.

7. Travis WD, Brambilla E, Noguchi M, et al. Diagnosis of lung adenocar-cinoma in resected specimens: implications of the 2011 International Associationfor the Study of Lung Cancer/American Thoracic Society/European RespiratorySociety classification. Arch Pathol Lab Med. 2013;137(5):685–705.

8. Goldstraw P, Crowley J, Chansky K, et al. The IASLC Lung Cancer StagingProject: proposals for the revision of the TNM stage groupings in the forthcoming(seventh) edition of the TNM classification of malignant tumours. J Thorac Oncol.2007;2(8):706–714.

9. Azzoli CG, Baker S Jr, Temin S, et al. American Society of ClinicalOncology Clinical Practice Guideline update on chemotherapy for stage IV non-small-cell lung cancer. J Clin Oncol. 2009;27(36):6251–6266.

10. Azzoli CG, Temin S, Aliff T, et al. 2011 focused update of 2009 AmericanSociety of Clinical Oncology Clinical Practice Guideline update on chemother-apy for stage IV non-small-cell lung cancer. J Clin Oncol. 2011;29(28):3825–3831.

11. Bonomi M, Pilotto S, Milella M, et al. Adjuvant chemotherapy forresected non-small-cell lung cancer: future perspectives for clinical research. JExp Clin Cancer Res. 2011;30:115.

12. Delbaldo C, Michiels S, Syz N, Soria JC, Le Chevalier T, Pignon JP.Benefits of adding a drug to a single-agent or a 2-agent chemotherapy regimen inadvanced non-small-cell lung cancer: a meta-analysis. JAMA. 2004;292(4):470–484.

13. Heon S, Johnson BE. Adjuvant chemotherapy for surgically resected non-small cell lung cancer. J Thorac Cardiovasc Surg. 2012;144(3):S39–S42.

14. Novello S, Milella M, Tiseo M, et al. Maintenance therapy in NSCLC:why? to whom? which agent? J Exp Clin Cancer Res. 2011;30(1):50.

15. Paoletti L, Pastis NJ, Denlinger CE, Silvestri GA. A decade of advances intreatment of early-stage lung cancer. Clin Chest Med. 2011;32(4):827–838.

16. Saintigny P, Burger JA. Recent advances in non-small cell lung cancerbiology and clinical management. Discov Med. 2012;13(71):287–297.

17. Sobol RE, Astarita RW, Hofeditz C, et al. Epidermal growth factor receptorexpression in human lung carcinomas defined by a monoclonal antibody. J NatlCancer Inst. 1987;79(3):403–407.

18. Lynch TJ, Bell DW, Sordella R, et al. Activating mutations in the epidermalgrowth factor receptor underlying responsiveness of non-small-cell lung cancerto gefitinib. N Engl J Med. 2004;350(21):2129–2139.

19. Paez JG, Janne PA, Lee JC, et al. EGFR mutations in lung cancer:correlation with clinical response to gefitinib therapy. Science. 2004;304(5676):1497–1500.

20. Pao W, Miller V, Zakowski M, et al. EGF receptor gene mutations arecommon in lung cancers from ‘‘never smokers’’ and are associated withsensitivity of tumors to gefitinib and erlotinib. Proc Natl Acad Sci U S A. 2004;101(36):13306–13311.

21. Mok TS, Wu YL, Thongprasert S, et al. Gefitinib or carboplatin-paclitaxelin pulmonary adenocarcinoma. N Engl J Med. 2009;361(10):947–957.

22. Mitsudomi T, Morita S, Yatabe Y, et al. Gefitinib versus cisplatin plusdocetaxel in patients with non-small-cell lung cancer harbouring mutations of theepidermal growth factor receptor (WJTOG3405): an open label, randomisedphase 3 trial. Lancet Oncol. 2010;11(2):121–128.

23. Maemondo M, Inoue A, Kobayashi K, et al. Gefitinib or chemotherapy fornon-small-cell lung cancer with mutated EGFR. N Engl J Med. 2010;362(25):2380–2388.

24. Rekhtman N, Paik PK, Arcila ME, et al. Clarifying the spectrum of driveroncogene mutations in biomarker-verified squamous carcinoma of lung: lack ofEGFR/KRAS and presence of PIK3CA/AKT1 mutations. Clin Cancer Res. 2012;18(4):1167–1176.

25. Cagle PT, Allen TC. Lung cancer genotype-based therapy and predictivebiomarkers: present and future. Arch Pathol Lab Med. 2012;136(12):1482–1491.

26. Chirieac LR, Dacic S. Targeted therapies in lung cancer. Surg Pathol Clin.2010;3(1):71–82.

27. Gaughan EM, Costa DB. Genotype-driven therapies for non-small celllung cancer: focus on EGFR, KRAS and ALK gene abnormalities. Ther Adv MedOncol. 2011;3(3):113–125.

28. Soda M, Choi YL, Enomoto M, et al. Identification of the transformingEML4-ALK fusion gene in non-small-cell lung cancer. Nature. 2007;448(7153):561–566.

29. Togashi Y, Soda M, Sakata S, et al. KLC1-ALK: a novel fusion in lungcancer identified using a formalin-fixed paraffin-embedded tissue only. PLoSOne. 2012;7(2):e31323.

30. Shaw AT, Yeap BY, Mino-Kenudson M, et al. Clinical features andoutcome of patients with non-small-cell lung cancer who harbor EML4-ALK. JClin Oncol. 2009;27(26):4247–4253.

31. Shaw AT, Yeap BY, Solomon BJ, et al. Effect of crizotinib on overall survivalin patients with advanced non-small-cell lung cancer harbouring ALK generearrangement: a retrospective analysis. Lancet Oncol. 2011;12(11):1004–1012.

1196 Arch Pathol Lab Med—Vol 137, September 2013 Lung Cancer Biomarkers: Present and Future—Cagle et al

Page 7: Ca Paru1

32. Shaw AT, Solomon B, Kenudson MM. Crizotinib and testing for ALK. JNatl Compr Canc Netw. 2011;9(12):1335–1341.

33. Bang YJ. The potential for crizotinib in non-small cell lung cancer: aperspective review. Ther Adv Med Oncol. 2011;3(6):279–291.

34. Katayama R, Shaw AT, Khan TM, et al. Mechanisms of acquired crizotinibresistance in ALK-rearranged lung cancers. Sci Transl Med. 2012;4(120):120ra17.

35. Zhang Z, Lee JC, Lin L, et al. Activation of the AXL kinase causesresistance to EGFR-targeted therapy in lung cancer. Nat Genet. 2012. 44(8):852–860.

36. Tochigi N, Dacic S, Nikiforova M, Cieply KM, Yousem SA. Adenosqua-mous carcinoma of the lung: a microdissection study of KRAS and EGFRmutational and amplification status in a Western patient population. Am J ClinPathol. 2011;135(5):783–789.

37. Gottschling S, Schnabel PA, Herth FJ, Herpel E. Are we missing the target:cancer stem cells and drug resistance in non-small cell lung cancer. CancerGenomics Proteomics. 2012;9(5):275–286.

38. Ettinger DS, Akerley W, Bepler G, et al. Non-small cell lung cancer. J NatlCompr Canc Netw. 2010;8(7):740–801.

39. Travis WD, Brambilla E, Noguchi M, et al. International Association forthe Study of Lung Cancer/American Thoracic Society/European RespiratorySociety: international multidisciplinary classification of lung adenocarcinoma. JThorac Oncol. 2011;6(2):244–285.

40. Richardson F, Richardson K, Sennello G, et al. Biomarker analysis fromcompletely resected NSCLC patients enrolled in an adjuvant erlotinib clinicaltrial (RADIANT). J Clin Oncol. 2010;27(15s):7520.

41. Janjigian YY, Park BJ, Kris MG, et al. Impact on disease-free survival ofadjuvant erlotinib or gefitinib in patients with resected lung adenocarcinomasthat harbor epidermal growth factor receptor (EGFR) mutations. J Clin Oncol.2009;27(15s):7523.

42. Neal JW, Pennell NA, Goodgame BW, et al. A multicenter phase II trial ofadjuvant erlotinib in patients with resected non-small cell lung cancer (NSCLC)and mutations in the epidermal growth factor receptor (EGFR): toxicityevaluation. J Clin Oncol. 2010;28(15s):abstract 7078.

43. Eng C. The evolving role of monoclonal antibodies in colorectal cancer:early presumptions and impact on clinical trial development. Oncologist. 2010;15(1):73–84.

44. Mehra R, Cohen RB, Burtness BA. The role of cetuximab for the treatmentof squamous cell carcinoma of the head and neck. Clin Adv Hematol Oncol.2008;6(10):742–750.

45. Moon C, Chae YK, Lee J. Targeting epidermal growth factor receptor inhead and neck cancer: lessons learned from cetuximab. Exp Biol Med(Maywood). 2010;235(8):907–920.

46. Vincenzi B, Zoccoli A, Pantano F, Venditti O, Galluzzo S. Cetuximab:from bench to bedside. Curr Cancer Drug Targets. 2010;10(1):80–95.

47. Wong SF. Cetuximab: an epidermal growth factor receptor monoclonalantibody for the treatment of colorectal cancer. Clin Ther. 2005;27(6):684–694.

48. Pirker R, Pereira JR, von Pawel J, et al. EGFR expression as a predictor ofsurvival for first-line chemotherapy plus cetuximab in patients with advancednon-small-cell lung cancer: analysis of data from the phase 3 FLEX study. LancetOncol. 2012;13(1):33–42.

49. Pirker R, Pereira JR, Szczesna A, et al. Prognostic factors in patients withadvanced non-small cell lung cancer: data from the phase III FLEX study. LungCancer. 2012;77(2):376–382.

50. O’Byrne KJ, Gatzemeier U, Bondarenko I, et al. Molecular biomarkers innon-small-cell lung cancer: a retrospective analysis of data from the phase 3 FLEXstudy. Lancet Oncol. 2011;12(8):795–805.

51. Ruschoff J, Kerr KM, Grote HJ, et al. Reproducibility of immunohisto-chemical scoring for epidermal growth factor receptor expression in non-smallcell lung cancer [published online ahead of print December 27, 2012]. ArchPathol Lab Med. doi:10.5858/arpa.2012-0605-OA.

52. Cagle PT, Olsen RJ. Verifying the role of surgical pathologists in theprecision medicine of lung cancer [published online ahead of print December31, 2012]. Arch Pathol Lab Med. doi:10.5858/arpa.2012-0659-ED.

53. Brugger W, Thomas M. EGFR-TKI resistant non-small cell lung cancer(NSCLC): new developments and implications for future treatment. Lung Cancer.2012;77(1):2–8.

54. Hirsch FR, Bunn PA Jr. A new generation of EGFR tyrosine-kinaseinhibitors in NSCLC. Lancet Oncol. 2012;13(5):442–443.

55. Langer CJ, Mok T, Postmus PE. Targeted agents in the third-/fourth-linetreatment of patients with advanced (stage III/IV) non-small cell lung cancer(NSCLC). Cancer Treat Rev. 2013;39(3):252–260.

56. Yang JC, Shih JY, Su WC, et al. Afatinib for patients with lungadenocarcinoma and epidermal growth factor receptor mutations (LUX-lung 2):a phase 2 trial. Lancet Oncol. 2012;13(5):539–548.

57. Ramalingam SS, Blackhall F, Krzakowski M, et al. Randomized phase IIstudy of dacomitinib (PF-00299804), an irreversible pan-human epidermalgrowth factor receptor inhibitor, versus erlotinib in patients with advanced non-small-cell lung cancer. J Clin Oncol. 2012;30(27):3337–3344.

58. Pietanza MC, Lynch TJ Jr, Lara PN Jr, et al. XL647—a multitargetedtyrosine kinase inhibitor: results of a phase II study in subjects with non-small celllung cancer who have progressed after responding to treatment with eithergefitinib or erlotinib. J Thorac Oncol. 2012;7(1):219–226.

59. Takeuchi K, Soda M, Togashi Y, et al. RET, ROS1 and ALK fusions in lungcancer. Nat Med. 2012;18(3):378–381.

60. Yasuda H, de Figueiredo-Pontes LL, Kobayashi S, Costa DB. Preclinicalrationale for use of the clinically available multitargeted tyrosine kinase inhibitorcrizotinib in ROS1-translocated lung cancer. J Thorac Oncol. 2012;7(7):1086–1090.

61. Janne PA, Meyerson M. ROS1 rearrangements in lung cancer: a newgenomic subset of lung adenocarcinoma. J Clin Oncol. 2012;30(8):878–879.

62. Reungwetwattana T, Molina JR, Mandrekar SJ, et al. Brief report: a phase II‘‘window-of-opportunity’’ frontline study of the mTOR inhibitor, temsirolimusgiven as a single agent in patients with advanced NSCLC, an NCCTG study. JThorac Oncol. 2012;7(5):919–922.

63. Xu CX, Li Y, Yue P, et al. The combination of RAD001 and NVP-BEZ235exerts synergistic anticancer activity against non-small cell lung cancer in vitroand in vivo. PLoS One. 2011;6(6):e20899.

64. Zito CR, Jilaveanu LB, Anagnostou V, et al. Multi-level targeting of thephosphatidylinositol-3-kinase pathway in non-small cell lung cancer cells. PLoSOne. 2012;7(2):e31331.

65. Zou ZQ, Zhang LN, Wang F, Bellenger J, Shen YZ, Zhang XH. The noveldual PI3K/mTOR inhibitor GDC-0941 synergizes with the MEK inhibitor U0126in non-small cell lung cancer cells. Mol Med Rep. 2012;5(2):503–508.

66. Sierra JR, Tsao MS. c-MET as a potential therapeutic target and biomarkerin cancer. Ther Adv Med Oncol. 2011;3(1 suppl):S21–S35.

67. Scagliotti GV, Novello S, Schiller JH, et al. Rationale and design ofMARQUEE: a phase III, randomized, double-blind study of tivantinib pluserlotinib versus placebo plus erlotinib in previously treated patients with locallyadvanced or metastatic, nonsquamous, non-small-cell lung cancer. Clin LungCancer. 2012;13(5):391–395.

68. Sequist LV, von Pawel J, Garmey EG, et al. Randomized phase II study oferlotinib plus tivantinib versus erlotinib plus placebo in previously treated non-small-cell lung cancer. J Clin Oncol. 2011;29(24):3307–3315.

69. Achcar Rde O, Cagle PT, Jagirdar J. Expression of activated and latentsignal transducer and activator of transcription 3 in 303 non-small cell lungcarcinomas and 44 malignant mesotheliomas: possible role for chemotherapeuticintervention. Arch Pathol Lab Med. 2007;131(9):1350–1360.

70. Shimokawa T, Seike M, Soeno C, et al. Enzastaurin has anti-tumour effectsin lung cancers with overexpressed JAK pathway molecules. Br J Cancer. 2012;106(5):867–875.

71. Zhang X, Yue P, Page BD, et al. Orally bioavailable small-moleculeinhibitor of transcription factor Stat3 regresses human breast and lung cancerxenografts. Proc Natl Acad Sci U S A. 2012;109(24):9623–9628.

72. Tanizaki J, Okamoto I, Takezawa K, et al. Combined effect of ALK andMEK inhibitors in EML4-ALK-positive non-small-cell lung cancer cells. Br JCancer. 2012;106(4):763–767.

73. Dy G. MEK/MAPK inhibitors. J Thorac Oncol. 2010;5(12 suppl 6):S474–S475.

74. Haura EB, Ricart AD, Larson TG, et al. A phase II study of PD-0325901,an oral MEK inhibitor, in previously treated patients with advanced non-small celllung cancer. Clin Cancer Res. 2010;16(8):2450–2457.

75. Kohno T, Ichikawa H, Totoki Y, et al. KIF5B-RET fusions in lungadenocarcinoma. Nat Med. 2012;18(3):375–377.

76. Ju YS, Lee WC, Shin JY, et al. A transforming KIF5B and RET gene fusion inlung adenocarcinoma revealed from whole-genome and transcriptome sequenc-ing. Genome Res. 2012;22(3):436–445.

77. Kruser TJ, Traynor AM, Wheeler DL. The use of single-agent dasatinib inmolecularly unselected non-small-cell lung cancer patients. Expert Opin InvestigDrugs. 2011;20(2):305–307.

78. Johnson ML, Riely GJ, Rizvi NA, et al. Phase II trial of dasatinib forpatients with acquired resistance to treatment with the epidermal growth factorreceptor tyrosine kinase inhibitors erlotinib or gefitinib. J Thorac Oncol. 2011;6(6):1128–1131.

79. Adjei AA. K-ras as a target for lung cancer therapy. J Thorac Oncol. 2008;3(6 suppl 2):S160–S163.

80. Cancer Genome Atlas Research Network; Hammerman PS, LawrenceMS, Voet D, et al. Comprehensive genomic characterization of squamous celllung cancers. Nature. 2012;489(7417):519–525.

81. Drilon A, Rekhtman N, Ladanyi M, Paik P. Squamous-cell carcinomas ofthe lung: emerging biology, controversies, and the promise of targeted therapy.Lancet Oncol. 2012;13(10):e418–e426.

82. Paik PK, Varghese AM, Sima CS, et al. Response to erlotinib in patientswith EGFR mutant advanced non-small cell lung cancers with a squamous orsquamous-like component. Mol Cancer Ther. 2012;11(11):2535–2540.

83. Perez-Moreno P, Brambilla E, Thomas R, Soria JC. Squamous cellcarcinoma of the lung: molecular subtypes and therapeutic opportunities. ClinCancer Res. 2012;18(9):2443–2451.

84. Schmid K, Bago-Horvath Z, Berger W, et al. Dual inhibition of EGFR andmTOR pathways in small cell lung cancer. Br J Cancer. 2010;103(5):622–628.

85. Sher T, Dy GK, Adjei AA. Small cell lung cancer. Mayo Clin Proc. 2008;83(3):355–367.

86. Tarhini A, Kotsakis A, Gooding W, et al. Phase II study of everolimus(RAD001) in previously treated small cell lung cancer. Clin Cancer Res. 2010;16(23):5900–5907.

87. Carney DN, Gazdar AF, Bunn PA Jr, Guccion JG. Demonstration of thestem cell nature of clonogenic tumor cells from lung cancer patients. Stem Cells.1982;1(3):149–164.

88. Shien K, Toyooka S, Ichimura K, et al. Prognostic impact of cancer stemcell-related markers in non-small cell lung cancer patients treated with inductionchemoradiotherapy. Lung Cancer. 2012;77(1):162–167.

Arch Pathol Lab Med—Vol 137, September 2013 Lung Cancer Biomarkers: Present and Future—Cagle et al 1197

Page 8: Ca Paru1

89. Alamgeer M, Peacock CD, Matsui W, Ganju V, Watkins DN. Cancer stemcells in lung cancer: evidence and controversies [published online ahead of printApril 16, 2013]. Respirology. doi:10.1111/resp.12094.

90. Gadepalli VS, Vaughan C, Rao RR. Isolation and characterization ofmurine multipotent lung stem cells. Methods Mol Biol. 2013;962:183–191.

91. Nurwidya F, Murakami A, Takahashi F, Takahashi K. Lung cancer stemcells: tumor biology and clinical implications. Asia Pac J Clin Oncol. 2012;8(3):217–222.

92. Lundin A, Driscoll B. Lung cancer stem cells: progress and prospects.Cancer Lett. 2012;pii:S0304-3835(12)00485-5.

93. Akunuru S, James Zhai Q, Zheng Y. Non-small cell lung cancer stem/progenitor cells are enriched in multiple distinct phenotypic subpopulations andexhibit plasticity. Cell Death Dis. 2012;3:e352.

94. Huang CP, Tsai MF, Chang TH, et al. ALDH-positive lung cancer stemcells confer resistance to epidermal growth factor receptor tyrosine kinaseinhibitors. Cancer Lett. 2013;328(1):144–151.

95. Yeh CT, Wu AT, Chang PM, et al. Trifluoperazine, an antipsychotic agent,inhibits cancer stem cell growth and overcomes drug resistance of lung cancer.Am J Respir Crit Care Med. 2012;186(11):1180–1188.

96. Gottschling S, Granzow M, Kuner R, et al. Mesenchymal stem cells innon-small cell lung cancer—different from others: insights from comparativemolecular and functional analyses. Lung Cancer. 2013;80(1):19–29.

97. Tsiatis AC, Norris-Kirby A, Rich RG, et al. Comparison of Sangersequencing, pyrosequencing, and melting curve analysis for the detection ofKRAS mutations: diagnostic and clinical implications. J Mol Diagn. 2010;12(4):425–432.

98. Lindeman N. Molecular diagnostics of lung cancers at the Brigham andWomen’s Hospital and Dana-Farber Cancer Institute: technology in rapidevolution. Arch Pathol Lab Med. 2012;136(10):1198–1200.

99. Cagle PT, Olsen RJ. The proposed new classification of pulmonaryadenocarcinoma and the conservation of small tissue samples for testing. ArchPathol Lab Med. 2013;137(4):453–454.

100. Dunlap J, Beadling C, Warrick A, et al. Multiplex high-throughput genemutation analysis in acute myeloid leukemia. Hum Pathol. 2012;43(12):2167–2176.

101. Yousem SA, Dacic S, Nikiforov YE, Nikiforova M. Pulmonary Langerhanscell histiocytosis: profiling of multifocal tumors using next-generation sequencingidentifies concordant occurrence of BRAF V600E mutations. Chest. 2013;143(6):1679–1684.

102. Rothberg JM, Leamon JH. The development and impact of 454sequencing. Nat Biotechnol. 2008;26(10):1117–1124.

103. Rothberg JM, Hinz W, Rearick TM, et al. An integrated semiconductordevice enabling non-optical genome sequencing. Nature. 2011;475(7356):348–352.

104. Tang K, Fu DJ, Julien D, Braun A, Cantor CR, Koster H. Chip-basedgenotyping by mass spectrometry. Proc Natl Acad Sci U S A. 1999;96(18):10016–10020.

105. Sakai K, Okamoto I, Takezawa K, et al. A novel mass spectrometry-basedassay for diagnosis of EML4-ALK-positive non-small cell lung cancer. J ThoracOncol. 2012;7(5):913–918.

106. Eid J, Fehr A, Gray J, et al. Real-time DNA sequencing from singlepolymerase molecules. Science. 2009;323(5910):133–138.

107. Loman NJ, Misra RV, Dallman TJ, et al. Performance comparison ofbenchtop high-throughput sequencing platforms. Nat Biotechnol. 2012;30(5):434–439.

108. Liu L, Li Y, Li S, et al. Comparison of next-generation sequencing systems.J Biomed Biotechnol. 2012;2012:251364.

109. Gargis AS, Kalman L, Berry MW, et al. Assuring the quality of next-generation sequencing in clinical laboratory practice. Nat Biotechnol. 2012;30(11):1033–1036.

110. McDermott U, Downing JR, Stratton MR. Genomics and the continuumof cancer care. N Engl J Med. 2011;364(4):340–350.

111. Imielinski M, Cha S, Rejtar T, Richardson EA, Karger BL, Sgroi DC.Integrated proteomic, transcriptomic, and biological network analysis of breastcarcinoma reveals molecular features of tumorigenesis and clinical relapse. MolCell Proteomics. 2012;11(6):M111.014910.

112. Gonzalez de Castro D, Clarke PA, Al-Lazikani B, Workman P.Personalized cancer medicine: molecular diagnostics, predictive biomarkers,and drug resistance. Clin Pharmacol Ther. 2013;93(3):252–259.

113. Brevet M, Arcila M, Ladanyi M. Assessment of EGFR mutation status inlung adenocarcinoma by immunohistochemistry using antibodies specific to thetwo major forms of mutant EGFR. J Mol Diagn. 2010;12(2):169–176.

114. Hasanovic A, Ang D, Moreira AL, Zakowski MF. Use of mutation specificantibodies to detect EGFR status in small biopsy and cytology specimens of lungadenocarcinoma. Lung Cancer. 2012;77(2):299–305.

115. Yu J, Kane S, Wu J, et al. Mutation-specific antibodies for the detection ofEGFR mutations in non-small-cell lung cancer. Clin Cancer Res. 2009;15(9):3023–3028.

116. Kwak EL, Bang YJ, Camidge DR, et al. Anaplastic lymphoma kinaseinhibition in non-small-cell lung cancer. N Engl J Med. 2010;363(18):1693–1703.

117. Boland JM, Erdogan S, Vasmatzis G, et al. Anaplastic lymphoma kinaseimmunoreactivity correlates with ALK gene rearrangement and transcriptionalup-regulation in non-small cell lung carcinomas. Hum Pathol. 2009;40(8):1152–1158.

118. Mino-Kenudson M, Chirieac LR, Law K, et al. A novel, highly sensitiveantibody allows for the routine detection of ALK-rearranged lung adenocarcino-mas by standard immunohistochemistry. Clin Cancer Res. 2010;16(5):1561–1571.

119. Yi ES, Boland JM, Maleszewski JJ, et al. Correlation of IHC and FISH forALK gene rearrangement in non-small cell lung carcinoma: IHC score algorithmfor FISH. J Thorac Oncol. 2011;6(3):459–465.

120. Cagle PT, Myers J. Precision medicine for lung cancer: role of the surgicalpathologist. Arch Pathol Lab Med. 2012;136(10):1186–1189.

121. Ross W, Lynch P, Raju G, et al. Biomarkers, bundled payments, andcolorectal cancer care. Genes Cancer. 2012;3(1):16–22.

122. Montero AJ, Avancha K, Gluck S, Lopes G. A cost-benefit analysis ofbevacizumab in combination with paclitaxel in the first-line treatment ofpatients with metastatic breast cancer. Breast Cancer Res Treat. 2012;132(2):747–751.

123. Schneider JE, Sidnu MK, Doucet C, Kiss N, Ohsfeld RL, Chalfin D.Economics of cancer biomarkers. Personalized Med. 2012;9(8):829–837.

1198 Arch Pathol Lab Med—Vol 137, September 2013 Lung Cancer Biomarkers: Present and Future—Cagle et al