Abstract

Personalized medicine relies upon the successful identification and translation of predictive biomarkers. Unfortunately, biomarker development has often fallen short of expectations. To better understand the obstacles to successful biomarker development, we systematically mapped research activities for a biomarker that has been in development for at least 12 years: excision repair cross‐complement group 1 protein (ERCC1) as a biomarker for predicting clinical benefit with platinum‐based chemotherapy in non‐small cell lung cancer. We found that although research activities explored a wide range of approaches to ERCC1 testing, there was little replication or validation of techniques, and design and reporting of results were generally poor. Our analysis points to problems with coordinating and standardizing research in biomarker development. Clinically meaningful progress in personalized medicine will require concerted efforts to address these problems. In the interim, health care providers should be aware of the complexity involved in biomarker development, cautious about their near‐term clinical value, and conscious of applying only validated diagnostics in the clinic.

Implications for Practice

Many hospitals, policy makers, and scientists have made ambitious claims about the promise of personalizing cancer care. When one uses a case example of excision repair cross‐complement group 1 protein—a biomarker that has a strong biological rationale and that has been researched for 12 years—the current research environment seems poorly suited for efficient development of biomarker tests. The findings provide grounds for tempering expectations about personalized cancer care—at least in the near term—and shed light on the current gap between the promise and practice of personalized medicine.

Introduction

The movement into the personalized medicine era brings much promise for cancer care. This is especially true for treatment modalities that are highly burdensome or that entail narrow therapeutic indices. Here, treatment decisions based on biomarker status—that is, prospectively testable and clinically informative properties of a patient’s biological material—might spare patients the burden of toxic and ineffective therapy.

But despite advances in understanding the mechanisms of tumor survival, development of clinically useful biomarkers for predicting response to cancer therapies has proven challenging [1]. Progress toward a diagnostic for the excision repair cross‐complement group 1 (ERCC1) protein as a predictive biomarker for platinum‐therapy in advanced non‐small cell lung cancer (NSCLC) provides a prototypical illustration of the obstacles blocking the transformation of molecular insights into clinically useful applications.

Platinum‐based doublet chemotherapy is a pillar of advanced NSCLC treatment. Cisplatin and carboplatin, the two most commonly used platinum therapies, work by adding platinum adducts to individual nucleotides. This prevents DNA replication, resulting in cancer cell death. The ERCC1 protein is the rate‐limiting factor in the nucleotide excision repair (NER) pathway, which removes platinum adducts as part of normal cellular activity [2]. Because expression levels of ERCC1 vary from patient to patient, this makes it an attractive marker for potentially explaining differences in patient outcomes after platinum‐based treatment [3].

During a decade of clinical research, dozens of studies have investigated the prospect of correlating ERCC1 with clinical outcomes in advanced NSCLC patients after platinum therapy. Yet at best, only modest progress has been made. To better understand why, we systematically mapped the research portfolio and evolution of evidence in support of ERCC1 testing.

Materials and Methods

Literature Search

We searched Embase and Medline databases to identify studies that investigated a relationship between ERCC1 expression and clinical outcomes in advanced NSCLC patients receiving platinum therapy. Search terms included cancer type (“non‐small cell lung cancer,” “NSCLC”), biomarker (“excision repair cross‐complement group 1,” “ERCC1,” or “ERCC‐1”), and the therapy (“platinum,” “cisplatin,” “carboplatin”). We excluded nonempirical and non‐English reports. The database results were then supplemented with a hand search based on reviews and study references . All studies were screened first by title and abstract, and then by full text. Exclusion criteria at screening included (a) meta‐analysis, (b) abstract only (c) not original data, (d) study population (i.e., not advanced stage), and (e) did not report objective response (OR) or overall survival (OS) data.

Data Extraction

Data extraction was implemented with the Numbat Extraction Framework (available for free from http://github.com/bgcarlisle/Numbat) and included the following domains: (a) study characteristics, (b) design, (c) assay properties, (d) reporting quality, (e) OR data and whether marker status was significantly associated with OS, and (f) the stated utility of the marker as predictive or prognostic. All reports were independently extracted by Barsanti‐Innes and Hey, and all discrepancies were settled through deliberation. Following the recommendations by Simon et al. (2009) [8], we created a five‐point scale for study quality based on (a) use of control specimens, (b) successful determination of biomarker status for more than 66% of the specimens in study sample, (c) assessment of biomarker status blinded to clinical data, and (d) prospective definition for biomarker‐positive status.

Patterns of Research Activity

To assess the organization of research activities, we used Accumulating Evidence and Research Organization (AERO) graphing [9]. This is a graph‐theoretic method for visually representing the trajectory and structure of research in a scientific domain. Briefly, an AERO graph represents each experiment as a node arranged on the x‐axis by time and stratified by study properties on the y‐axis. Nodes are then color‐coded to represent the evolution of evidence. For our AERO analysis, we stratified by five levels: (a) assay method, (b) tissue type, (c) assay reagents, (d) prespecified cutoff value, and (e) drug regimen.

Results

Sample Characteristics

Twenty‐eight studies met our eligibility criteria; Figure 1 shows the PRISMA flow diagram. These reports spanned 12 years of published research; properties of our sample are shown in Table 1  . This study sample reflects 4,311 patient samples (2,295 tested for protein expression and 2,016 for mRNA) in total. Only 14% of the studies received financial support from commercial sponsors.

Preferred reporting items for systematic reviews and meta‐analyses (PRISMA) flow diagram.
Figure 1

Preferred reporting items for systematic reviews and meta‐analyses (PRISMA) flow diagram.

Abbreviation: ERCC1, excision repair cross‐complement group 1 protein.

Table 1

Basic study characteristics

CharacteristicTrials (N =  28), % (n)
Study type
Prospective clinical trial7 (2)
Retrospective/biobank study93 (26)
Sponsor
Nonindustry72 (20)
Industry14 (4)
Not stated14 (4)
Number of centers
Single center79 (22)
Multicenter21 (6)
Assays used
Immunohistochemistry54 (15)
Quantitative reverse‐transcriptase polymerase chain reaction43 (12)
AQUA3 (1)
Methodological quality
Use of control specimens79 (22)
>66% specimens successfully analyzed71 (20)
Definition for biomarker‐positive status64 (18)
Outcome assessment blinded to biomarker status61 (17)
Self‐reported trend for ERCC1 as useful biomarker
Positive75 (21)
Negative25 (7)
Location of corresponding author
Asia57 (16)
Europe29 (8)
North America11 (3)
Africa3 (1)
CharacteristicTrials (N =  28), % (n)
Study type
Prospective clinical trial7 (2)
Retrospective/biobank study93 (26)
Sponsor
Nonindustry72 (20)
Industry14 (4)
Not stated14 (4)
Number of centers
Single center79 (22)
Multicenter21 (6)
Assays used
Immunohistochemistry54 (15)
Quantitative reverse‐transcriptase polymerase chain reaction43 (12)
AQUA3 (1)
Methodological quality
Use of control specimens79 (22)
>66% specimens successfully analyzed71 (20)
Definition for biomarker‐positive status64 (18)
Outcome assessment blinded to biomarker status61 (17)
Self‐reported trend for ERCC1 as useful biomarker
Positive75 (21)
Negative25 (7)
Location of corresponding author
Asia57 (16)
Europe29 (8)
North America11 (3)
Africa3 (1)

Abbreviations: AQUA, automated quantitative analysis; ERCC1, excision repair cross‐complement group 1.

Table 1

Basic study characteristics

CharacteristicTrials (N =  28), % (n)
Study type
Prospective clinical trial7 (2)
Retrospective/biobank study93 (26)
Sponsor
Nonindustry72 (20)
Industry14 (4)
Not stated14 (4)
Number of centers
Single center79 (22)
Multicenter21 (6)
Assays used
Immunohistochemistry54 (15)
Quantitative reverse‐transcriptase polymerase chain reaction43 (12)
AQUA3 (1)
Methodological quality
Use of control specimens79 (22)
>66% specimens successfully analyzed71 (20)
Definition for biomarker‐positive status64 (18)
Outcome assessment blinded to biomarker status61 (17)
Self‐reported trend for ERCC1 as useful biomarker
Positive75 (21)
Negative25 (7)
Location of corresponding author
Asia57 (16)
Europe29 (8)
North America11 (3)
Africa3 (1)
CharacteristicTrials (N =  28), % (n)
Study type
Prospective clinical trial7 (2)
Retrospective/biobank study93 (26)
Sponsor
Nonindustry72 (20)
Industry14 (4)
Not stated14 (4)
Number of centers
Single center79 (22)
Multicenter21 (6)
Assays used
Immunohistochemistry54 (15)
Quantitative reverse‐transcriptase polymerase chain reaction43 (12)
AQUA3 (1)
Methodological quality
Use of control specimens79 (22)
>66% specimens successfully analyzed71 (20)
Definition for biomarker‐positive status64 (18)
Outcome assessment blinded to biomarker status61 (17)
Self‐reported trend for ERCC1 as useful biomarker
Positive75 (21)
Negative25 (7)
Location of corresponding author
Asia57 (16)
Europe29 (8)
North America11 (3)
Africa3 (1)

Abbreviations: AQUA, automated quantitative analysis; ERCC1, excision repair cross‐complement group 1.

Methodological Quality

Two of 28 studies in our cohort (7%) used a prospective biomarker trial design. The others were retrospective analyses. Pathologists were named as coauthors in 68% of studies. The sample size of each study was generally small, ranging from 35 to 443 with a median of 101 specimen samples per study. Mean quality score was 2.6. Twelve investigations (43%) reported no more than two of our four quality‐score elements. Use of controls and the proportion of samples successfully analyzed were inadequately reported in 21% and 29% of studies, respectively. A prospective cutoff for biomarker status was unreported in 36% of studies, and blinding of pathologists determining biomarker status to clinical response went unreported in 39% of studies. We did not observe an obvious trend toward greater rigor in study design as studies advanced from hypothesis generation toward confirmatory testing. No study after 2010 adequately reported all four methodological practices. Also, fewer than half of studies discussed the analytical validity of their biomarker diagnostics (12 of 28; 43%).

Predictive Versus Prognostic Biomarker Potential

Most of the studies (82%) included only patients who had received platinum therapy. However, 7 of those 23 (35%) made claims of testing predictive marker utility, 3 (11%) made claims of testing prognostic utility, and 18 (64%) made a claim that conflated the prognostic and predictive utility of the marker.

Patterns of Research Activity

Tables 2 and 3 list the diversity of components used in the study designs of our sample. Even simple biomarker tests consist of a coordinated set of practices and conditions, including a definition of tissues to be analyzed, an assay method, and a scoring rule. We call these coordinated practices and conditions a “biomarker ensemble.” Figure 2 depicts all of the combinations of the five components defining our biomarker ensemble that are present in our sample. In total, 24 different combinations were investigated. However, only three of these combinations were ever replicated.

Accumulating Evidence and Research Organization graph stratified by biomarker ensemble, as defined by assay, tissue type, reagents, prespecified cutoff value, and drug regimen. Square nodes are retrospective analyses, and circular nodes are prospective trials. Numbers within nodes represent the sample size. Green nodes are studies that showed a statistically significant association between low excision repair cross‐complement group 1 (ERCC1) expression on both objective response rate and overall survival. Yellow nodes are studies that showed statistical significance for one outcome but not for the other. Red nodes showed no statistically significant associations. The gray node showed an opposite association—that is, high ERCC1 expression was associated with better response and survival. Red shading indicates investigations that fell below a quality score of 3. Pragmatic regimen is patient population treated with more than two regimens.
Figure 2

Accumulating Evidence and Research Organization graph stratified by biomarker ensemble, as defined by assay, tissue type, reagents, prespecified cutoff value, and drug regimen. Square nodes are retrospective analyses, and circular nodes are prospective trials. Numbers within nodes represent the sample size. Green nodes are studies that showed a statistically significant association between low excision repair cross‐complement group 1 (ERCC1) expression on both objective response rate and overall survival. Yellow nodes are studies that showed statistical significance for one outcome but not for the other. Red nodes showed no statistically significant associations. The gray node showed an opposite association—that is, high ERCC1 expression was associated with better response and survival. Red shading indicates investigations that fell below a quality score of 3. Pragmatic regimen is patient population treated with more than two regimens.

Abbreviations: car, carboplatin; cis, cisplatin; doc, docetaxel; gem, gemcitabine; ifo, ifosfamide; iri, irinotecan; NS, not stated; pac, paclitaxel; pem, pemetrexed; Prim./Meta., primary/metastatic.

Table 2

Characteristics of excision repair cross‐complement group 1 biomarker studies using protein expression assays

AuthorYearTypeCutoffTherapyNOROSTissue sampleQ
Wachters et al. [10]2005Retro>10% stainingCis + gem37NoNoEither4
Fuji et al. [11]2008RetroMdn HCis + iri, cis + doc35YesNoMetastatic4
Hwang et al. [26]2008RetroMdn HCis + pac/doc/etop, car + pac/doc83NoYesMetastatic4
Lee et al. [12]2009RetroMdn HCis + gem/doc/vin, car + gem/pac/doc51NoYesEither4
Ota et al. [29]2009Retro>10% stainingCis + vin/doc/iri/gem, car + pac200NoYesNS4
Jeong et al. [27]2010RetroMdn HCis + etop/ifo/doc77NoNoNS3
Vilmar et al. [33]2010RetroMdn HCis + pac/gem/vin443NoYesEither3
Wang et al. [34]2010Retro>10% stainingCis + gem/vin/doc/145YesYesPrimary4
Bepler et al. [13]2013Phase III66.0Car + gem/doc275NoNoNS2
Lee et al. [28]2013RetroMdn HCis + pem41YesYesNS3
Ozdemir et al. [14]2013RetroMdn Hcis/car + gem/etop/vin/doc/pac/pem/mit83NoNoNS2
Tiseo et al. [15]2013RetroMdn HCis + gem, cis + gem + ifo433NoYesPrimary0
Vassalou et al. [16]2013RetroMdn Hcis‐based, car‐based94NoYesEither3
Yamashita et al. [35]2013RetroMdn scoreCis + vin/doc/gem/pem/etop, car + pac/gem/pem/vin103NoNoNS2
Yan et al. [17]2013RetroMdn HCis + gem/pac115NRYesPrimary3
Sad et al. [31]2014Retro>10% stainingcis + gem/pac, car + pac80YesYesNS1
AuthorYearTypeCutoffTherapyNOROSTissue sampleQ
Wachters et al. [10]2005Retro>10% stainingCis + gem37NoNoEither4
Fuji et al. [11]2008RetroMdn HCis + iri, cis + doc35YesNoMetastatic4
Hwang et al. [26]2008RetroMdn HCis + pac/doc/etop, car + pac/doc83NoYesMetastatic4
Lee et al. [12]2009RetroMdn HCis + gem/doc/vin, car + gem/pac/doc51NoYesEither4
Ota et al. [29]2009Retro>10% stainingCis + vin/doc/iri/gem, car + pac200NoYesNS4
Jeong et al. [27]2010RetroMdn HCis + etop/ifo/doc77NoNoNS3
Vilmar et al. [33]2010RetroMdn HCis + pac/gem/vin443NoYesEither3
Wang et al. [34]2010Retro>10% stainingCis + gem/vin/doc/145YesYesPrimary4
Bepler et al. [13]2013Phase III66.0Car + gem/doc275NoNoNS2
Lee et al. [28]2013RetroMdn HCis + pem41YesYesNS3
Ozdemir et al. [14]2013RetroMdn Hcis/car + gem/etop/vin/doc/pac/pem/mit83NoNoNS2
Tiseo et al. [15]2013RetroMdn HCis + gem, cis + gem + ifo433NoYesPrimary0
Vassalou et al. [16]2013RetroMdn Hcis‐based, car‐based94NoYesEither3
Yamashita et al. [35]2013RetroMdn scoreCis + vin/doc/gem/pem/etop, car + pac/gem/pem/vin103NoNoNS2
Yan et al. [17]2013RetroMdn HCis + gem/pac115NRYesPrimary3
Sad et al. [31]2014Retro>10% stainingcis + gem/pac, car + pac80YesYesNS1

The OR/OS columns indicate significant association found between marker‐positive status and either tumor response or overall survival. Abbreviations: car, carboplatin; cis, cisplatin; doc, docetaxel; etop, etoposide; gem, gemcitabine; H, H score; ifo, ifosfamide; iri, irinotecan; Mdn, median; mit, mitomycin; N, number of participants recruited into the study; NR, not reported; NS, not stated; OR, objective response; OS, overall survival; pac, paclitaxel; pem, pemetrexed; Q, 5‐point (0–4) quality score; Retro, retrospective; vin, vinorelbine.

Table 2

Characteristics of excision repair cross‐complement group 1 biomarker studies using protein expression assays

AuthorYearTypeCutoffTherapyNOROSTissue sampleQ
Wachters et al. [10]2005Retro>10% stainingCis + gem37NoNoEither4
Fuji et al. [11]2008RetroMdn HCis + iri, cis + doc35YesNoMetastatic4
Hwang et al. [26]2008RetroMdn HCis + pac/doc/etop, car + pac/doc83NoYesMetastatic4
Lee et al. [12]2009RetroMdn HCis + gem/doc/vin, car + gem/pac/doc51NoYesEither4
Ota et al. [29]2009Retro>10% stainingCis + vin/doc/iri/gem, car + pac200NoYesNS4
Jeong et al. [27]2010RetroMdn HCis + etop/ifo/doc77NoNoNS3
Vilmar et al. [33]2010RetroMdn HCis + pac/gem/vin443NoYesEither3
Wang et al. [34]2010Retro>10% stainingCis + gem/vin/doc/145YesYesPrimary4
Bepler et al. [13]2013Phase III66.0Car + gem/doc275NoNoNS2
Lee et al. [28]2013RetroMdn HCis + pem41YesYesNS3
Ozdemir et al. [14]2013RetroMdn Hcis/car + gem/etop/vin/doc/pac/pem/mit83NoNoNS2
Tiseo et al. [15]2013RetroMdn HCis + gem, cis + gem + ifo433NoYesPrimary0
Vassalou et al. [16]2013RetroMdn Hcis‐based, car‐based94NoYesEither3
Yamashita et al. [35]2013RetroMdn scoreCis + vin/doc/gem/pem/etop, car + pac/gem/pem/vin103NoNoNS2
Yan et al. [17]2013RetroMdn HCis + gem/pac115NRYesPrimary3
Sad et al. [31]2014Retro>10% stainingcis + gem/pac, car + pac80YesYesNS1
AuthorYearTypeCutoffTherapyNOROSTissue sampleQ
Wachters et al. [10]2005Retro>10% stainingCis + gem37NoNoEither4
Fuji et al. [11]2008RetroMdn HCis + iri, cis + doc35YesNoMetastatic4
Hwang et al. [26]2008RetroMdn HCis + pac/doc/etop, car + pac/doc83NoYesMetastatic4
Lee et al. [12]2009RetroMdn HCis + gem/doc/vin, car + gem/pac/doc51NoYesEither4
Ota et al. [29]2009Retro>10% stainingCis + vin/doc/iri/gem, car + pac200NoYesNS4
Jeong et al. [27]2010RetroMdn HCis + etop/ifo/doc77NoNoNS3
Vilmar et al. [33]2010RetroMdn HCis + pac/gem/vin443NoYesEither3
Wang et al. [34]2010Retro>10% stainingCis + gem/vin/doc/145YesYesPrimary4
Bepler et al. [13]2013Phase III66.0Car + gem/doc275NoNoNS2
Lee et al. [28]2013RetroMdn HCis + pem41YesYesNS3
Ozdemir et al. [14]2013RetroMdn Hcis/car + gem/etop/vin/doc/pac/pem/mit83NoNoNS2
Tiseo et al. [15]2013RetroMdn HCis + gem, cis + gem + ifo433NoYesPrimary0
Vassalou et al. [16]2013RetroMdn Hcis‐based, car‐based94NoYesEither3
Yamashita et al. [35]2013RetroMdn scoreCis + vin/doc/gem/pem/etop, car + pac/gem/pem/vin103NoNoNS2
Yan et al. [17]2013RetroMdn HCis + gem/pac115NRYesPrimary3
Sad et al. [31]2014Retro>10% stainingcis + gem/pac, car + pac80YesYesNS1

The OR/OS columns indicate significant association found between marker‐positive status and either tumor response or overall survival. Abbreviations: car, carboplatin; cis, cisplatin; doc, docetaxel; etop, etoposide; gem, gemcitabine; H, H score; ifo, ifosfamide; iri, irinotecan; Mdn, median; mit, mitomycin; N, number of participants recruited into the study; NR, not reported; NS, not stated; OR, objective response; OS, overall survival; pac, paclitaxel; pem, pemetrexed; Q, 5‐point (0–4) quality score; Retro, retrospective; vin, vinorelbine.

Table 3

Characteristics of excision repair cross‐complement group 1 biomarker studies using mRNA assays

AuthorYearTypeCutoffTherapyNOROSTissue sampleQ
Lord et al. [18]2002RetroMdnCis + gem56NoYesPrimary3
Ceppi et al. [19]2006RetroMdnCis + gem70NoYesNS3
Booton et al. [20]2007RetroMdnCis + mit + ifo/vin, car + doc108NoNoNS2
Simon et al. [21]2007Phase II8.7Car + gem/doc75NoNoEither4
Ren et al. [22]2010RetroMdnCis/car + gem/vin/pac/doc100NoYesNS3
Joerger et al. [23]2011RetroNRCis/car + gem137YesYesNS2
Su et al. [32]2011RetroMdnCis/car + gem/vin/pac130NoYesNS3
Zhang et al. [24]2012RetroMdnCar + gem52NoYesNS tumor + peripheral2
Jian‐Wei et al. [25]2013RetroMdnCis/car + gem/vin/pac294YesYesPeripheral2
Qiao et al. [30]2014RetroMdnCis + gem305YesYesPeripheral1
Wang et al. [37]2014RetroMdnThird‐generation platinum + gem/vin/pac366YesYesPeripheral1
Zhang et al. [36]2014RetroMdnCis/car + gem/vin/pac323YesYesPeripheral1
AuthorYearTypeCutoffTherapyNOROSTissue sampleQ
Lord et al. [18]2002RetroMdnCis + gem56NoYesPrimary3
Ceppi et al. [19]2006RetroMdnCis + gem70NoYesNS3
Booton et al. [20]2007RetroMdnCis + mit + ifo/vin, car + doc108NoNoNS2
Simon et al. [21]2007Phase II8.7Car + gem/doc75NoNoEither4
Ren et al. [22]2010RetroMdnCis/car + gem/vin/pac/doc100NoYesNS3
Joerger et al. [23]2011RetroNRCis/car + gem137YesYesNS2
Su et al. [32]2011RetroMdnCis/car + gem/vin/pac130NoYesNS3
Zhang et al. [24]2012RetroMdnCar + gem52NoYesNS tumor + peripheral2
Jian‐Wei et al. [25]2013RetroMdnCis/car + gem/vin/pac294YesYesPeripheral2
Qiao et al. [30]2014RetroMdnCis + gem305YesYesPeripheral1
Wang et al. [37]2014RetroMdnThird‐generation platinum + gem/vin/pac366YesYesPeripheral1
Zhang et al. [36]2014RetroMdnCis/car + gem/vin/pac323YesYesPeripheral1

The OR and OS columns indicate significant association found between marker‐positive status and either tumor response or overall survival.

aIn contrast to every other report, this study found a significant improvement in response rate and overall survival in patients with high excision repair cross‐complement group 1 expression.

Abbreviations: car, carboplatin; cis, cisplatin; doc, docetaxel; gem, gemcitabine; ifo, ifosfamide; Mdn, median; mit, mitomycin; N, number of participants recruited into the study; NR, none reported; NS, not stated; OR, objective response; OS, overall survival; pac, paclitaxel; Q, 5‐point (0–4) quality score; Retro, retrospective; vin, vinorelbine.

Table 3

Characteristics of excision repair cross‐complement group 1 biomarker studies using mRNA assays

AuthorYearTypeCutoffTherapyNOROSTissue sampleQ
Lord et al. [18]2002RetroMdnCis + gem56NoYesPrimary3
Ceppi et al. [19]2006RetroMdnCis + gem70NoYesNS3
Booton et al. [20]2007RetroMdnCis + mit + ifo/vin, car + doc108NoNoNS2
Simon et al. [21]2007Phase II8.7Car + gem/doc75NoNoEither4
Ren et al. [22]2010RetroMdnCis/car + gem/vin/pac/doc100NoYesNS3
Joerger et al. [23]2011RetroNRCis/car + gem137YesYesNS2
Su et al. [32]2011RetroMdnCis/car + gem/vin/pac130NoYesNS3
Zhang et al. [24]2012RetroMdnCar + gem52NoYesNS tumor + peripheral2
Jian‐Wei et al. [25]2013RetroMdnCis/car + gem/vin/pac294YesYesPeripheral2
Qiao et al. [30]2014RetroMdnCis + gem305YesYesPeripheral1
Wang et al. [37]2014RetroMdnThird‐generation platinum + gem/vin/pac366YesYesPeripheral1
Zhang et al. [36]2014RetroMdnCis/car + gem/vin/pac323YesYesPeripheral1
AuthorYearTypeCutoffTherapyNOROSTissue sampleQ
Lord et al. [18]2002RetroMdnCis + gem56NoYesPrimary3
Ceppi et al. [19]2006RetroMdnCis + gem70NoYesNS3
Booton et al. [20]2007RetroMdnCis + mit + ifo/vin, car + doc108NoNoNS2
Simon et al. [21]2007Phase II8.7Car + gem/doc75NoNoEither4
Ren et al. [22]2010RetroMdnCis/car + gem/vin/pac/doc100NoYesNS3
Joerger et al. [23]2011RetroNRCis/car + gem137YesYesNS2
Su et al. [32]2011RetroMdnCis/car + gem/vin/pac130NoYesNS3
Zhang et al. [24]2012RetroMdnCar + gem52NoYesNS tumor + peripheral2
Jian‐Wei et al. [25]2013RetroMdnCis/car + gem/vin/pac294YesYesPeripheral2
Qiao et al. [30]2014RetroMdnCis + gem305YesYesPeripheral1
Wang et al. [37]2014RetroMdnThird‐generation platinum + gem/vin/pac366YesYesPeripheral1
Zhang et al. [36]2014RetroMdnCis/car + gem/vin/pac323YesYesPeripheral1

The OR and OS columns indicate significant association found between marker‐positive status and either tumor response or overall survival.

aIn contrast to every other report, this study found a significant improvement in response rate and overall survival in patients with high excision repair cross‐complement group 1 expression.

Abbreviations: car, carboplatin; cis, cisplatin; doc, docetaxel; gem, gemcitabine; ifo, ifosfamide; Mdn, median; mit, mitomycin; N, number of participants recruited into the study; NR, none reported; NS, not stated; OR, objective response; OS, overall survival; pac, paclitaxel; Q, 5‐point (0–4) quality score; Retro, retrospective; vin, vinorelbine.

The precise content of these combinations is further complicated by parameters that have mixed or unreported populations. Of the studies in our sample, 39% enrolled patients from more than two different treatment regimens, which we classified as “pragmatic” regimens. Also, 39% of studies did not specify the type of tissue used for analysis, largely rendering these results uninterpretable. In each of the three replicated ensembles at least one parameter had such a mixed population (tissue type, pragmatic regimen, or both).

The two prospective studies in our sample applied diagnostic components that had been not been previously validated—the 18SrNA internal reference gene [21] and automated quantitative analysis technique [13]. These particular techniques were then never followed up in later research.

Only six studies in our sample used a prespecified numerical cutoff (i.e., did not use the median score in the sample population as the cutoff). However, two of the six studies that reported an externally valid cutoff also used a pragmatic patient population , and one did not specify the location of the tumor sample [13].

Finally, in 50% of the studies in our sample there was discordance between ERCC1’s predictive correlation with OR and OS (represented by yellow nodes in Fig. 2). Even where these outcomes were concordant within a study, there was no consistency across our sample. Twenty‐five percent of studies reported concordant positive outcomes, and 25% reported concordant negative outcomes.

Discussion

The state of uncertainty regarding the value of ERCC1 testing is, in many ways, representative of personalized medicine more generally [1]. Many commentators have cataloged impediments to the successful translation of biomarkers, extending from validity issues in basic science [38] to the large‐scale coordination of geographically and technologically disparate research centers . Accordingly, there are numerous sets of recommendations in the literature for improving the analytical validity of assay reagents and increasing necessary reporting for tumor marker studies . There have also been calls for implementing a registry system focused on tumor markers and establishing a system with clear milestones for determining biomarker utility .

Our analysis suggests other impediments as well. Diagnostic tests consist of a set of practices—such as assay protocols, tumor preservation methods, type of tissues analyzed, scoring criteria, and so on—that must be combined to unlock the clinical value of a given biomarker. Determining whether a biomarker diagnostic should be implemented for patient management requires optimizing each of these variables [48]. The ERCC1 biomarker translation trajectory shows how difficult and prolonged translation can be when poor study design and reporting combines with a haphazard approach to testing these different variables.

For example, of the 16 studies in our sample that measured ERCC1 protein expression, all but one of them used an antibody, 8F1, that may not exclusively target ERCC1. The antibody 8F1 first appears in our cohort in 2005 and is utilized for the next 9 years, despite concerns relating to its specificity emerging in 2007 [49]. One study showed that ERCC1 is not the principal antigen recognized by 8F1 and that this antibody is incapable of distinguishing between high and low ERCC1 expression in human cell lines [50]. More recently, ERCC1 protein has been shown to exist in four functionally distinct isoforms, with only one of them displaying the target NER capacity [51]. Given that stratification of marker‐positive and ‐negative groups may have been based on faulty reagents, this may mean that half of our sample produced uninformative results.

Similar validity issues confront studies evaluating mRNA. Reports in our sample do not appear to have clearly addressed the selection of the internal reference gene used to quantify the amount of ERCC1 mRNA. Only one study noted that they used a reference gene that had confirmed normal expression in tumor cells [20]. Also, the primers used during the quantitative reverse‐ transcriptase polymerase chain reaction process are not isoform specific, resulting in the same potential for misclassification that may have occurred with the protein evaluations [6].

Scoring criteria represent another dimension that confounds predictive marker validation. Of the studies in our sample, 75% used the median value of ERCC1 expression to stratify their patient populations in a binary fashion according to whether they exceeded or were lower than this value. Although specifying a median value cutoff may be useful in exploratory investigations, this approach lacks the capacity for externally valid inference and compromises development of a useful patient selection criterion for use in a clinical setting.

Controversy also exists about the comparability of molecular profiles between tumor sites . Studies contrasting ERCC1 protein expression between primary and metastatic tissues in NSCLC have shown that levelsare often unrelated . Similarly, ERCC1 mRNA levels have been shown to lack the correlation between tumor and peripheral samples [24]. Despite this, 18% of the studies in our sample combined results from patient specimens that came from disparate sites, and 46% did not report the location of the sample origin. Clinicians wishing to apply personalized medicine strategies—or advance the science—need to be aware that how and from where biospecimens are collected can dramatically influence the results of biomarker analyses.

Variation in patient drug regimen is another potential confounder and barrier to progress. Cisplatin and carboplatin, the two most common types of platinum therapy, while known to target DNA using similar mechanisms, are also known to have different toxicity and efficacy profiles [57]. Yet half of the studies in our sample did not limit patient eligibility on the basis of which drug they received. This further complicates the ability of researchers to isolate the effect of ERCC1 expression on the response to a particular treatment.

NSCLC platinum therapy is also typically given in combination with other drugs—often as a doublet with paclitaxel, gemcitabine, docetaxel, vinorelbine, irinotecan, or pemetrexed [58]. Of the studies in our sample, 79% compared patients who had been treated with different drug regimens, and 39% compared more than two different regimens. This introduces another potential confound in isolating the effect of ERCC1 in moderating the effectiveness of the platinum therapy.

Biomarkers can also serve different purposes in directing patient management. A predictive marker identifies a population that is more likely to respond favorably to a particular therapy, thereby directly influencing patient treatment selection. In contrast a purely prognostic marker means that when treatment is not taken into consideration, a biomarker‐selected population will have a better clinical outcome. Markers can be prognostic or predictive or have characteristics of both. To distinguish between these two types of markers, a study must compare patient data from both biomarker groups, in which sufficient patients from each designation receive the marker‐associated therapy as well as standard of care, as in a randomized clinical trial. Only five studies in our cohort included ERCC1 high‐ and low‐expressing patients and compared the difference in outcomes between platinum‐based and nonplatinum‐based regimens. The remaining 23 studies included only patients who received platinum therapy, making it difficult to determine a truly predictive or prognostic use for the ERCC1 diagnostic. This information is necessary to guide clinicians tasked with selecting appropriate patient treatments .

Finally, the low‐level of reporting quality, particularly among the more recent investigations, represents unethical research waste of scarce research and biospecimen resources. As Figure 2 shows, 12 studies failed to report more than two of our four methodological elements. This greatly compromises the interpretability of reports and renders findings of little use in systematic reviews or quantitative synthesis.

The difficulties being experienced by researchers evaluating ERCC1 as a biomarker for personalizing medicine has, unfortunately, not prevented the introduction of several commercially available diagnostics promoting the ability to use the marker in selecting patients for platinum therapy . A recent study evaluating the reliability of three such assays demonstrated large variability between the tests in determining ERCC1 expression levels in identical patient samples, and furthermore, none could distinguish responders from nonresponders to platinum therapy [66]. Furthermore, clinical guidelines have vacillated in their inclusion of ERCC1 as a useful biomarker. In 2011 a special National Comprehensive Cancer Network task force reported ERCC1 as a biomarker with emerging evidence [67], and in 2013 their clinical practice guidelines included it as a marker to predict the efficacy of platinum therapy in NSCLC patients [68]. However, the most recent guidelines make no reference to the marker [69]. Yet several of the tests remain commercially available.

Our analysis of the ERCC1 research program illustrates how, despite a wide exploration of biomarker ensemble parameters and very compelling biological and clinical rationales, there has been little or no convergence on an optimal, clinically useful technique for stratifying patients on the basis of ERCC1 marker status. This can be explained, in part, by a failure to address the many dimensions of uncertainty surrounding a biomarker test in a systematic fashion. In the ideal development trajectory, a range of plausible values for the key elements in the ensemble would be tested in an exploratory setting. Once optimal values are discovered, these would then be incorporated into large‐ scale, decisive tests in appropriate patient populations to determine the diagnostic’s utility.

The current, passive mechanisms for research coordination appear insufficient to motivate and support attainment of key translation milestones of optimizing each component. This underscores the need for novel social mechanisms to guide activities in this domain, both in the form of a top‐down regulatory body that can help direct investigators toward critical unanswered questions and in the form of better adherence to standards in methodology and reporting practices. One possibility for biomarker research moving forward is the establishment of a formalized tool for coordinating ongoing investigations. In our work we present the AERO diagram as a means of retrospectively illustrating past issues in the portfolio of ERCC1 research. However, by tracking the progressing accumulation and reporting quality of research, it could also provide a valuable prospective tool to improve biomarker development. In highlighting unexplored areas of research, or areas of conflict, an AERO diagram would help direct investigators to the most pressing areas of research. Particularly if this tool were given the support of agencies such as the National Cancer Institute or the National Human Genome Research Institute, this kind of official status could incentivize its use among individual researchers, improve research communication, and reduce research waste.

Furthermore, there is an important gatekeeping role for journal publishers and peer reviewers in vetting submitted articles to ensure their adherence to reporting guidelines such as REMARK (REporting recommendations for tumour MARKer prognostic studies). Clinicians and especially pathologists participating in biomarker research either directly or through the provision of biospecimens can play a role in this by being aware of past and present research in their area to ensure that resources are being used efficiently to answer the most pressing translational questions and by ensuring that their own publications, and those that they review, meet reporting standards.

Our study has some limitations. Our literature search will have excluded some studies that examined the assay methods in isolation from the particular clinical application of interest. We also note that our “quality scale” has not been validated as a metric. However, we do not think it likely that these limitations, if rectified, would substantially alter our analysis or conclusions.

Conclusion

Researchers, policy makers, and funding agencies often talk about biomarkers as though their predictive capacities reside in a particular molecular signature rather than the clinical and assay operations used to access and define that signature . Simple “biomarker speak” can obscure the challenges of both translating and implementing personalized cancer care.

Our findings reinforce three points that bear on the practice of personalized cancer care. First, clinicians should be mindful that the prognostic or predictive value of a biomarker is highly sensitive to the conditions under which a biomarker test is implemented. Realizing the promise of personalized cancer care requires testing and optimizing each of these conditions. Second, current research systems are not well suited to efficient and reliable testing of these conditions. Research on a biomarker is often pursued in parallel rather than in a coordinated fashion, greatly prolonging development. Physicians can advocate for a more methodical and coordinated approach to research. Finally, an awareness of the scientific and social complexities in translating prognostic or predictive biomarkers should motivate a more cautious appraisal of the near‐term potential of personalized medicine.

Acknowledgments

This study was conducted under the PACEOMICS project, funded by Genome Canada, Genome Quebec, Genome Alberta, and the Canadian Institute for Health Research.

Author Contributions

Conception/Design: Brianna Barsanti‐Innes, Spencer Phillips Hey, Jonathan Kimmelman

Collection and/or assembly of data: Brianna Barsanti‐Innes, Spencer Phillips Hey, Jonathan Kimmelman

Data analysis and interpretation: Brianna Barsanti‐Innes, Spencer Phillips Hey, Jonathan Kimmelman

Manuscript writing: Brianna Barsanti‐Innes, Spencer Phillips Hey, Jonathan Kimmelman

Final approval of manuscript: Brianna Barsanti‐Innes, Spencer Phillips Hey, Jonathan Kimmelman

Disclosures

The authors indicated no financial relationships.

References

1

Hayes
 
DF
,
Allen
 
J
,
Compton
 
C
 et al. .
Breaking a vicious cycle
.
Sci Transl Med
 
2013
;
5
:
196cm6
.

2

Altaha
 
R
,
Liang
 
X
,
Yu
 
JJ
 et al. .
Excision repair cross complementing‐group: 1. Gene expression and platinum resistance
.
Int J Mol Med
 
2004
;
14
:
959
970
.

3

Olaussen
 
KA
,
Dunant
 
A
,
Fouret
 
P
 et al. .
DNA repair by ERCC1 in non‐small‐cell lung cancer and cisplatin‐based adjuvant chemotherapy
.
N Engl J Med
 
2006
;
355
:
983
991
.

4

Bonanno
 
L
.
Predictive models for customizing chemotherapy in advanced non‐small cell lung cancer (NSCLC)
.
Transl Lung Cancer Res
 
2013
;
2
:
160
171
.

5

Hubner
 
RA
,
Riley
 
RD
,
Billingham
 
LJ
 et al. .
Excision repair cross‐complementation group 1 (ERCC1) status and lung cancer outcomes: A meta‐analysis of published studies and recommendations
.
PLoS One
 
2011
;
6
:
e25164
.

6

Besse
 
B
,
Olaussen
 
KA
,
Soria
 
JC
.
ERCC1 and RRM1: Ready for primetime?
 
J Clin Oncol
 
2013
;
31
:
1050
1060
.

7

Roth
 
JA
,
Carlson
 
JJ
.
Prognostic role of ERCC1 in advanced non‐small‐cell lung cancer: A systematic review and meta‐analysis
.
Clin Lung Cancer
 
2011
;
12
:
393
401
.

8

Simon
 
RM
,
Paik
 
S
,
Hayes
 
DF
.
Use of archived specimens in evaluation of prognostic and predictive biomarkers
.
J Natl Cancer Inst
 
2009
;
101
:
1446
1452
.

9

Hey
 
SP
,
Heilig
 
CM
,
Weijer
 
C
.
Accumulating evidence and research organization (AERO) model: A new tool for representing, analyzing, and planning a translational research program
.
Trials
 
2013
;
14
:
159
.

10

Wachters
 
FM
,
Wong
 
LS
,
Timens
 
W
 et al. .
ERCC1, hRad51, and BRCA1 protein expression in relation to tumour response and survival of stage III/IV NSCLC patients treated with chemotherapy
.
Lung Cancer
 
2005
;
50
:
211
219
.

11

Fujii
 
T
,
Toyooka
 
S
,
Ichimura
 
K
 et al. .
ERCC1 protein expression predictsthe response of cisplatin‐ based neoadjuvant chemotherapy in non‐small‐cell lung cancer
.
Lung Cancer
 
2008
;
59
:
377
384
.

12

Lee
 
HW
,
Choi
 
YW
,
Han
 
JH
 et al. .
Expression of excision repair cross‐complementation group 1 protein predicts poor outcome in advanced non small cell lung cancer patients treated with platinum‐ based doublet chemotherapy
.
Lung Cancer
 
2009
;
65
:
377
382
.

13

Bepler
 
G
,
Williams
 
C
,
Schell
 
MJ
 et al. .
Randomized international phase III trial of ERCC1 and RRM1 expression‐based chemotherapy versus gemcitabine/carboplatin in advanced non‐small‐cell lung cancer
.
J Clin Oncol
 
2013
;
31
:
2404
2412
.

14

Ozdemir
 
O
,
Ozdemir
 
P
,
Veral
 
A
 et al. .
ERCC1 expression does not predict survival and treatment response in advanced stage non‐small cell lung cancer cases treated with platinum based chemotherapy
.
Asian Pac J Cancer Prev
 
2013
;
14
:
4679
4683
.

15

Tiseo
 
M
,
Bordi
 
P
,
Bortesi
 
B
 et al. .
ERCC1/BRCA1 expression and gene polymorphisms as prognostic and predictive factors in advanced NSCLC treated with or without cisplatin
.
Br J Cancer
 
2013
;
108
:
1695
1703
.

16

Vassalou
 
H
,
Stathopoulos
 
E
,
Fiolitaki
 
G
 et al. .
Excision‐repair‐cross‐complement‐1 protein as a prognostic factor in patients with advanced nonsmall cell lung cancer treated with platinum‐based first‐line chemotherapy
.
Lung Cancer
 
2013
;
82
:
324
329
.

17

Yan
 
D
,
Wei
 
P
,
An
 
G
 et al. .
Prognostic potential of ERCC1 protein expression and clinicopathologic factors in stage III/N2 non‐small cell lung cancer
.
J Cardiothorac Surg
 
2013
;
8
:
149
.

18

Lord
 
RV
,
Brabender
 
J
,
Gandara
 
D
 et al. .
Low ERCC1 expression correlates with prolonged survival after cisplatin plus gemcitabine chemotherapy in non‐small cell lung cancer
.
Clin Cancer Res
 
2002
;
8
:
2286
2291
.

19

Ceppi
 
P
,
Volante
 
M
,
Novello
 
S
 et al. .
ERCC1 and RRM1 gene expressions but not EGFRare predictive of shorter survival in advanced non‐small‐cell lung cancer treated with cisplatin and gemcitabine
.
Ann Oncol
 
2006
;
17
:
1818
1825
.

20

Booton
 
R
,
Ward
 
T
,
Ashcroft
 
L
 et al. .
ERCC1 mRNA expression is not associated with response and survival after platinum‐based chemotherapy regimens in advanced non‐small cell lung cancer
.
J Thorac Oncol
 
2007
;
2
:
902
906
.

21

Simon
 
G
,
Sharma
 
A
,
Li
 
X
 et al. .
Feasibility and efficacy of molecular analysis‐directed individualized therapy in advanced non‐small‐cell lung cancer
.
J Clin Oncol
 
2007
;
25
:
2741
2746
.

22

Ren
 
S
,
Zhou
 
S
,
Zhang
 
L
 et al. .
High‐level mRNA of excision repair cross‐complementation group 1 gene is associated with poor outcome of platinum‐ based doublet chemotherapy of advanced nonsmall cell lung cancer patients
.
Cancer Invest
 
2010
;
28
:
1078
1083
.

23

Joerger
 
M
,
deJong
 
D
,
Burylo
 
A
 et al. .
Tubulin, BRCA1, ERCC1, Abraxas, RAP80 mRNA expression, p53/p21 immunohistochemistry and clinical outcome in patients with advanced non small‐cell lung cancer receiving first‐line platinum‐gemcitabine chemotherapy
.
Lung Cancer
 
2011
;
74
:
310
317
.

24

Zhang
 
GB
,
Chen
 
J
,
Wang
 
LR
 et al. .
RRM1 and ERCC1 expression in peripheral blood versus tumor tissue in gemcitabine/carboplatin‐treated advanced non‐small cell lung cancer
.
Cancer Chemother Pharmacol
 
2012
;
69
:
1277
1287
.

25

Jian‐Wei
 
B
,
Yi‐Min
 
M
,
Yu‐Xia
 
S
 et al. .
Expression levels of ERCC1 and RRM1 mRNA and clinical outcome of advanced non‐small cell lung cancer
.
Pak J Med Sci
 
2013
;
29
:
1158
1161
.

26

Hwang
 
IG
,
Ahn
 
MJ
,
Park
 
BB
 et al. .
ERCC1 expression as a prognostic marker in N2(+) nonsmall‐cell lung cancer patients treated with platinum‐based neoadjuvant concurrent chemoradiotherapy
.
Cancer
 
2008
;
113
:
1379
1386
.

27

Jeong
 
SH
,
Jung
 
JH
,
Han
 
JH
 et al. .
Expression of Bcl‐2 predicts outcome in locally advanced nonsmall cell lungcancer patients treated with cisplatin‐based concurrent chemoradiotherapy
.
Lung Cancer
 
2010
;
68
:
288
294
.

28

Lee
 
SH
,
Noh
 
KB
,
Lee
 
JS
 et al. .
Thymidylate synthase and ERCC1 as predictive markers in patients with pulmonary adenocarcinoma treated with pemetrexed and cisplatin
.
Lung Cancer
 
2013
;
81
:
102
108
.

29

Ota
 
S
,
Ishii
 
G
,
Goto
 
K
 et al. .
Immunohistochemical expression of BCRP and ERCC1 in biopsy specimen predicts survival in advanced non‐small cell lung cancer treated with cisplatin‐based chemotherapy
.
Lung Cancer
 
2009
;
64
:
98
104
.

30

Qiao
 
H
,
Huang
 
X
,
Guo
 
H
 et al. .
ERCC1, RRM1 and TUBB3 mRNA expression on the tumor response and overall survival of non‐small cell lung cancer treated with platinum‐based chemotherapy
.
Pak J Med Sci
 
2014
;
30
:
1403
1408
.

31

Sad
 
LM
,
Younis
 
SG
,
Elity
 
MM
.
Prognostic and predictive role of ERCC1 protein expression in locally advanced stage III non‐small cell lung cancer
.
Med Oncol
 
2014
;
31
:
58
.

32

Su
 
C
,
Zhou
 
S
,
Zhang
 
L
 et al. .
ERCC1, RRM1 and BRCA1 mRNA expression levels and clinical outcome of advanced non‐small cell lung cancer
.
Med Oncol
 
2011
;
28
:
1411
1417
.

33

Vilmar
 
AC
,
Santoni‐Rugiu
 
E
,
Sorensen
 
JB
.
ERCC1 and histopathology in advanced NSCLC patients randomized in a large multicenter phase III trial
.
Ann Oncol
 
2010
;
21
:
1817
1824
.

34

Wang
 
X
,
Zhao
 
J
,
Yang
 
L
 et al. .
Positive expression of ERCC1 predicts a poorer platinum‐based treatment outcome in Chinese patients with advanced non‐small‐cell lung cancer
.
Med Oncol
 
2010
;
27
:
484
490
.

35

Yamashita
 
F
,
Azuma
 
K
,
Yoshida
 
T
 et al. .
Prognostic value of EGFR mutation and ERCC1 in patients with non‐small cell lungcancer undergoing platinum‐ based chemotherapy
.
PLoS One
 
2013
;
8
:
e71356
.

36

Zhang
 
H
,
Li
 
J
,
Zhang
 
Y
 et al. .
ERCC1 mRNA expression is associated with the clinical outcome of non‐small cell lung cancer treated with platinum‐ based chemotherapy
.
Genet Mol Res
 
2014
;
13
:
10215
10222
.

37

Wang
 
TB
,
Zhang
 
NL
,
Wang
 
SH
 et al. .
Expression of ERCC1 and BRCA1 predict the clinical outcome of non‐small cell lung cancer in patients receiving platinum‐based chemotherapy
.
Genet Mol Res
 
2014
;
13
:
3704
3710
.

38

Haibe‐Kains
 
B
,
El‐Hachem
 
N
,
Birkbak
 
NJ
 et al. .
Inconsistency in large pharmacogenomic studies
.
Nature
 
2013
;
504
:
389
393
.

39

de
 
Gramont
 
A
,
Watson
 
S
,
Ellis
 
LM
 et al. .
Pragmatic issues in biomarkere valuation for targeted the rapies in cancer
.
Nat Rev Clin Oncol
 
2015
;
12
:
197
212
.

40

McShane
 
LM
,
Cavenagh
 
MM
,
Lively
 
TG
 et al. .
Criteria for the use of omics‐based predictors in clinical trials
.
Nature
 
2013
;
502
:
317
320
.

41

Anagnostou
 
VK
,
Welsh
 
AW
,
Giltnane
 
JM
 et al. .
Analytic variability in immunohistochemistry biomarker studies
.
Cancer Epidemiol Biomarkers Prev
 
2010
;
19
:
982
991
.

42

Moore
 
HM
,
Kelly
 
AB
,
Jewell
 
SD
 et al. .
Biospecimen reporting for improved studyquality (BRISQ)
.
J Proteome Res
 
2011
;
10
:
3429
3438
.

43

McShane
 
LM
,
Altman
 
DG
,
Sauerbrei
 
W
 et al. .
Reporting recommendations for tumor marker prognostic studies (REMARK)
.
J Natl Cancer Inst
 
2005
;
97
:
1180
1184
.

44

Bordeaux
 
J
,
Welsh
 
A
,
Agarwal
 
S
 et al. .
Antibody validation
.
Biotechniques
 
2010
;
48
:
197
209
.

45

Andre
 
F
,
McShane
 
LM
,
Michiels
 
S
 et al. .
Biomarker studies: A call for a comprehensive biomarker study registry
.
Nat Rev Clin Oncol
 
2011
;
8
:
171
176
.

46

The WIN Biomarker Studies Registry
. Available at http://winbiomarkerstudies.com/.

47

Hayes
 
DF
,
Bast
 
RC
,
Desch
 
CE
 et al. .
Tumor marker utility grading system: A framework to evaluate clinical utility of tumor markers
.
J Natl Cancer Inst
 
1996
;
88
:
1456
1466
.

48

Hey
 
S
.
Judging quality and coordination in biomarker diagnostic development
.
Theoria
 
2015
;
30
:
207
227
.

49

Niedernhofer
 
LJ
,
Bhagwat
 
N
,
Wood
 
RD
.
ERCC1 and non‐small‐cell lung cancer
.
N Engl J Med
 
2007
;
356
:
2538
2540
.

50

Ma
 
D
,
Baruch
 
D
,
Shu
 
Y
 et al. .
Using protein micro array technology to screen anti‐ERCC1 monoclonal antibodies for specificity and applications in pathology
.
BMC Biotechnol
 
2012
;
12
:
88
.

51

Friboulet
 
L
,
Olaussen
 
KA
,
Pignon
 
JP
 et al. .
ERCC1 isoform expression and DNA repair in non‐small‐cell lung cancer
.
N Engl J Med
 
2013
;
368
:
1101
1110
.

52

Daniele
 
L
,
Cassoni
 
P
,
Bacillo
 
E
 et al. .
Epidermal growth factor receptor gene in primary tumor and metastatic sites from non‐small cell lung cancer
.
J Thorac Oncol
 
2009
;
4
:
684
688
.

53

Italiano
 
A
,
Vandenbos
 
FB
,
Otto
 
J
 et al. .
Comparison of the epidermal growth factor receptor gene and protein in primary non‐small‐cell‐lung cancer and metastatic sites: Implications for treatment with EGFR‐inhibitors
.
Ann Oncol
 
2006
;
17
:
981
985
.

54

Weigelt
 
B
,
Glas
 
AM
,
Wessels
 
LF
 et al. .
Gene expression profiles of primary breast tumors maintained in distant metastases
.
Proc Natl Acad Sci USA
 
2003
;
100
:
15901
15905
.

55

Kang
 
CH
,
Jang
 
BG
,
Kim
 
DW
 et al. .
Differences in the expression profiles of excision repair crosscomplementation group 1, x‐ray repair crosscomplementation group 1, and betaIII‐tubulin between primary non‐small cell lung cancer and metastatic lymph nodes and the significance in mid‐term survival
.
J Thorac Oncol
 
2009
;
4
:
1307
1312
.

56

Gomez‐Roca
 
C
,
Raynaud
 
CM
,
Penault‐Llorca
 
F
 et al. .
Differential expression of biomarkers in primary non‐small cell lung cancer and metastatic sites
.
J Thorac Oncol
 
2009
;
4
:
1212
1220
.

57

Vilmar
 
A
,
Sørensen
 
JB
.
Excision repair crosscomplementation group 1 (ERCC1) in platinum‐ based treatment of non‐small cell lung cancer with special emphasis on carboplatin: A review of current literature
.
Lung Cancer
 
2009
;
64
:
131
139
.

58

National Cancer Institute
.
Non‐Small Cell Lung Cancer Treatment (PDQ) Stage IV NSCLC Treatment
. Available at http://www.cancer.gov/cancertopics/pdq/treatment/non-small-cell-lung/healthprofessional/. Accessed October 22,
2015
.

59

Henry
 
NL
,
Hayes
 
DF
.
Uses and abuses of tumor markers in the diagnosis, monitoring, and treatment of primary and metastatic breast cancer
.
The Oncologist
 
2006
;
11
:
541
552
.

60

Ballman
 
KV
.
Biomarker: Predictive or prognostic?
 
J Clin Oncol
 
2015
;
33
:
3968
3971
.

61

ERCC1 Analysis in Non‐Small Cell Lung Cancer
. Available at http://www.integratedoncology.com/sites/default/files/Onc-757-v7-101315-Prognostic%20Therapeutic.pdf. Accessed October 22,
2015
.

62

Testing
 
P
.
Delivering Precision Medicine
. Available at http://www.cancergenetics.com/pharmacogenomics-home/pgx-testing/oncology/. Accessed October 22,
2015
.

63

ERCC1 Gene Expression Analysis Kit
. Available at http://www.mobitec.com/cms/products/bio/09_ivd/Real-Time_PCR_Cancer_Diagnostic_Kits.html?pdf=ADx-ER01.pdf. Accessed October 22,
2015
.

64

Quest Diagnostics
.
ERCC1, IHC With Interpretation
. Available at http://www.questdiagnostics.com/testcenter/TestDetail.action?ntc=16979. Accessed October 22,
2015
.

65

ERCC1
. Available at http://www.clarient.com/Test-Menu/ercc1. Accessed October 22,
2015
.

66

Schneider
 
JG
,
Farhadfar
 
N
,
Sivapiragasam
 
A
 et al. .
Commercial laboratory testing of excision repair cross‐complementation group 1 expression in non‐small cell lung cancer
.
The Oncologist
 
2014
;
19
:
459
465
.

67

Febbo
 
PG
,
Ladanyi
 
M
,
Aldape
 
KD
 et al. .
NCCN Task Force report: Evaluating the clinical utility of tumor markers in oncology
.
J Natl Compr Canc Netw
 
2011
;
9
(
suppl 5
):
S1
S32
.

68

Ettinger
 
DS
,
Akerley
 
W
,
Borghaei
 
H
 et al. .
Non‐small cell lung cancer, version 2.2013
.
J Natl Compr Canc Netw
 
2013
;
11
:
645
653
.

69

Ettinger
 
DS
,
Wood
 
DE
,
Akerley
 
W
 et al. .
NCCN guidelines insights: Non‐small cell lung cancer, version 4.2016
.
J Natl Compr Canc Netw
 
2016
;
14
:
255
264
.

Author notes

Disclosures of potential conflicts of interest may be found at the end of this article.

This article is published and distributed under the terms of the Oxford University Press, Standard Journals Publication Model (https://dbpia.nl.go.kr/journals/pages/open_access/funder_policies/chorus/standard_publication_model)