We use cookies to improve your experience. By continuing to browse this site, you accept our cookie policy.×
Skip main navigation
Aging Health
Bioelectronics in Medicine
Biomarkers in Medicine
Breast Cancer Management
CNS Oncology
Colorectal Cancer
Concussion
Epigenomics
Future Cardiology
Future Medicine AI
Future Microbiology
Future Neurology
Future Oncology
Future Rare Diseases
Future Virology
Hepatic Oncology
HIV Therapy
Immunotherapy
International Journal of Endocrine Oncology
International Journal of Hematologic Oncology
Journal of 3D Printing in Medicine
Lung Cancer Management
Melanoma Management
Nanomedicine
Neurodegenerative Disease Management
Pain Management
Pediatric Health
Personalized Medicine
Pharmacogenomics
Regenerative Medicine
Short CommunicationOpen Accesscc iconby iconnc iconnd icon

Evolution of RAS testing over time: factors influencing mutation rates in metastatic colorectal cancer patients

    Kelly Dufraing

    Biomedical Quality Assurance Research Unit, Department of Public Health & Primary Care, University of Leuven, Leuven, Belgium

    Authors contributed equally

    Search for more papers by this author

    ,
    Cleo Keppens

    Biomedical Quality Assurance Research Unit, Department of Public Health & Primary Care, University of Leuven, Leuven, Belgium

    Authors contributed equally

    Search for more papers by this author

    ,
    Véronique Tack

    Biomedical Quality Assurance Research Unit, Department of Public Health & Primary Care, University of Leuven, Leuven, Belgium

    ,
    Albert Gerrit Siebers

    Department of Pathology, Radboud University Medical Center, Nijmegen, The Netherlands

    ,
    George Kafatos

    Amgen Ltd, Uxbridge, United Kingdom

    ,
    Sabada Dube

    Amgen Ltd, Uxbridge, United Kingdom

    ,
    Gaston Demonty

    Amgen Belgium S.A./N.V., Brussels, Belgium

    ,
    Kimberly Lowe

    Amgen Inc, Thousand Oaks, CA 91320, USA

    ,
    Leonie Ilse Kroeze

    Department of Pathology, Radboud University Medical Center, Nijmegen, The Netherlands

    ,
    Marjolijn Ligtenberg

    Department of Pathology, Radboud University Medical Center, Nijmegen, The Netherlands

    Department of Human Genetics, Radboud University Medical Center, Nijmegen, The Netherlands

    ,
    Nicola Normanno

    Cell Biology & Biotherapy Unit, Istituto Nazionale Tumori – Fondazione Pascale, Naples, Italy

    ,
    Lien Tembuyser

    Biomedical Quality Assurance Research Unit, Department of Public Health & Primary Care, University of Leuven, Leuven, Belgium

    ,
    Vander Borght Sara

    Pathologische Ontleedkunde, Department of Pathology, University Hospitals Leuven, Belgium

    ,
    Joannes Henricus van Krieken

    Department of Pathology, Radboud University Medical Center, Nijmegen, The Netherlands

    &
    Elisabeth Marie C Dequeker

    *Author for correspondence: Tel.: +32 1634 5881;

    E-mail Address: els.dequeker@kuleuven.be

    Biomedical Quality Assurance Research Unit, Department of Public Health & Primary Care, University of Leuven, Leuven, Belgium

    Published Online:https://doi.org/10.2217/crc-2019-0013

    Abstract

    Aim: Correct identification of RAS gene variants is key for targeted treatment decisions in patients with metastatic colorectal cancer. Published RAS mutation rates differ and could be influenced by several factors including testing methods. This study aimed to describe the performance of laboratories to correctly identify RAS variants over time and to understand how RAS testing has evolved in Europe. Materials & methods: Misclassification and test failure rates were calculated and related to the used test methodology for 239 unique laboratories participating in external quality assessment for metastatic colorectal cancer between 2013 and 2018. In addition, 33 laboratories completed a survey aiming to obtain more details on their routine testing strategies, number of samples analyzed and RAS mutation rates between 2013 and 2017. Results: The mutation status was correctly analyzed in 96.1% (N = 5471) RAS and BRAF tests. A total of 4.6% (N = 2860) RAS tests included false-negative results. In 1.6% (N = 5562) RAS and BRAF tests, an analysis failure occurred. Misclassifications and technical failures both decreased between 2013 and 2018. The number of next-generation sequencing users increased from 6.9% (N = 130) in 2013 to 44.6% (N = 112) in 2018. Over time, more codons were included in the methodologies, yet 23.2% (N = 112) did not offer full RAS testing (exon 2, 3, 4) in 2018. Based on the survey the overall RAS mutation rate was estimated as 45.2% (N = 27,325). Conclusion: This is the largest observational study reporting RAS mutation rates to-date. There was no trend of RAS mutation rates over time despite having a clear shift to more sensitive tests and increased quality of testing.

    Approximately 1.85 million new cases of colorectal cancer (CRC) were diagnosed in 2018 and is the third most commonly diagnosed cancer worldwide, representing 10.2% of all cancers [1,2]. In newly diagnosed cases, 20–25% will have already developed metastasis and up to 50% of all CRC patients eventually develop metastatic disease [2–6]. The 5-year survival rate for patients with metastatic CRC (mCRC) is approximately 14% [7].

    Over the past 15 years, the introduction of monoclonal antibodies (mAbs), such as cetuximab and panitumumab into mCRC treatment has led to an improved overall and progression-free survival for many patients [8–12]. By binding to the EGFR, both cetuximab and panitumumab disrupt several cancerous signaling pathways involved in a variety of cellular processes such as control of cell growth and survival, cell cycle progression, cell proliferation and metastasis [13,14].

    Observational studies have initially shown that mCRC patients with KRAS mutations in exon 2 (codons 12 and 13) are less likely to benefit from anti-EGFR therapies [15–21]. Further studies identified a number of additional mutations in exons 2, 3 and 4 of KRAS and NRAS, which impact on response to anti-EGFR therapies [9,22,23]. Therefore, in July 2013, the therapeutic indication for panitumumab was restricted by the European Medicines Agency (EMA) to patients with RAS (exons 2, 3 and 4 of KRAS and NRAS) wild-type mCRC tumors only (codons 12, 13, 59, 61, 117, 146). In November 2013, the EMA also determined that cetuximab could only be given to mCRC patients without a RAS mutation

    Mutations in the BRAF gene occur in 6–15% of all CRC patients [24,25]. Testing for this gene is often combined with RAS testing for its prognostic value, rather than for predicting response to anti-EGFR mAbs, as there is no clear evidence of its predictive role at the moment [17,26].

    It is not well understood whether or how the label changes impacted RAS testing for mCRC. Laboratories may have had to broaden their testing extensiveness, introduce new methods of higher sensitivity or test a higher number of samples. In addition to testing practices, improvement of the quality of laboratory testing is important to understand trends in laboratory testing and RAS mutation rates.

    This study aimed to understand how RAS testing has evolved over time among mCRC patients in Europe and to evaluate whether laboratory performance is satisfactory 5 years after the EMA label changed. This manuscript consists of European-wide longitudinal data from two sources: external quality assessment (EQA) to evaluate misclassifications related to testing methodology; and a laboratory survey on the number of samples tested and RAS mutation rates in mCRC patients by methodology.

    Material & methods

    A retrospective observational study with two components was conducted: EQA data from the European Society of Pathology (ESP) Colon EQA scheme; and information was collected from a subset of laboratories participating in the ESP Colon schemes between 2013 and 2017 via a survey.

    EQA scheme set-up

    Data from the ESP Colon EQA schemes were collected between 2013 and 2018 according to ISO 17043 [27,28], from 239 unique laboratories from 44 different countries.

    Each participant received three glass slides from ten formalin-fixed paraffin embedded tissue samples for pathological review, DNA extraction and mutation analysis. The preparation and validation of the samples was performed by regional scheme organizers until 2016 [29] and by one reference laboratory from 2017, following confirmation by another reference laboratory with experience in molecular pathology. All sample material was remaining resection specimens from real-life patients, carefully validated for containing a minimum of 30% neoplastic cells and sufficient DNA quantity and quality for testing by the participants.

    Participants were asked to analyze these samples for RAS hotspot variants and optional BRAF hotspot variants using their routine procedures within 14 calendar days. In addition, participants provided information on the applied methodologies along with their testing results in an online datasheet. Results were assessed by a team of international experts in molecular pathology using pre-defined and harmonized scoring criteria [27].

    EQA data analysis

    For all analyses, only samples that contained neoplastic cells were included. In 2013, 18 tests were excluded from the analysis because the primer sequences used were unable to anneal to the template artificial DNA as a result of gene modulation technology. No samples from 2015 were included as there was not an EQA scheme organized in that year.

    Misclassification rates were calculated as the total number of incorrect results, divided by the total number of performed tests, for which an outcome could be provided. Incorrect results included false-positive (FP) results (identification of a variant in a wild-type case, or an additional variant besides the correct variant), false-negative (FN) results (variant in the sample unidentified) and wrong mutations that affected the mutation rate (variant in another gene reported resulting in an FN or FP results for the respective gene). Wrong mutations that did not influence the mutation rate or possible therapy decision (e.g., NRAS c.34G>C p.[Gly12Arg] reported, instead of NRAS c.34G>T p.[Gly12Cys]), were not included in the ‘misclassification rate' definition. The true FN rates were calculated as the total of FN results, for samples containing a variant (with the exclusion of WT cases). To calculate misclassification rates related to test methodology, only methods by which at least 50 tests were performed in all scheme years are included. Technical failures are defined as samples for which no outcome could be given because of a test failure. The failure rate was calculated as the total number of technical failures divided by the total number of tests. Laboratories participated successfully to the EQA program if they obtained a score of at least 90% and made no major genotyping error, in other words, errors which could possibly lead to the administration of the wrong therapy.

    Pathology data collection via a survey

    All European centers that participated in the ESP Colon EQA schemes between 2013 and 2017 were invited to complete an online survey, to collect data on testing strategies, the number of samples tested annually for mCRC and the annual RAS mutation rate. The survey questions and responses are shown in Supplementary Data 1.

    Average numbers of RAS samples tested per laboratory for a specific year were calculated from total numbers of RAS samples tested by all laboratories over the number of laboratories that provided information on that year. Only 6 months of data was available for years 2013 and 2017. To achieve comparable estimates per laboratory, the same testing rate was assumed for the remainder 6 months of these years, and hence, the means for these 2 years were doubled. The RAS mutation rate was calculated as the total number of samples of RAS mutant status over the samples with RAS known status:

    RAS mutation rate = RAS mutant samplesRAS mutant samples + RAS wild-type samples

    Results

    ESP Colon EQA schemes

    From all unique laboratories participating in the ESP Colon EQA schemes between 2013 and 2018, 41.0% (98/239) were situated in a university hospital, 26.8% (64/239) in a community hospital, 18.0% (43/239) were private laboratories, 7.1% (17/239) related to an anticancer institute, 5.0% (12/239) were situated in a private hospital and 2.1% (5/239) were laboratories from the industry (Table 1).

    Table 1. Overview of laboratory setting and testing performance in external quality assessment over time for varying extensiveness of RAS testing.
     20132014201620172018
     Full RASFull KRAS onlyIncomplete KRAS and NRASTotalFull RASFull KRAS onlyIncomplete KRAS and NRASTotalFull RASFull KRAS onlyIncomplete KRAS and NRASTotalFull RASFull KRAS onlyIncomplete KRAS and NRASTotalFull RASFull KRAS onlyIncomplete KRAS and NRASTotal
    Laboratory setting                    
    University hospital27 (47.4%)14 (24.6%)16
    (28.1%)
    57 (43.8%)38 (70.4%)8 (14.8%)8
    (14.8%)
    54 (43.5%)38 (76.0%)4 (8.0%)8
    (16.0%)
    50 (41.0%)35 (92.1%)2 (5.3%)1
    (2.6%)
    38 (35.5%)37 (80.4%)5 (10.9%)4
    (8.7%)
    46 (41.1%)
    Community hospital13 (37.1%)8 (22.9%)14
    (40.0%)
    35 (26.9%)26 (66.7%)6 (15.4%)7
    (17.9%)
    39 (31.5%)23 (65.7%)6 (17.1%)6
    (17.1%)
    35 (28.7%)29 (85.3%)1 (2.9%)4
    (11.8%)
    34 (31.8%)29 (82.9%)2 (5.7%)4
    (11.4%)
    35 (31.3%)
    Private hospital2 (25.0%)4 (50.0%)2
    (25.0%)
    8 (6.2%)5 (71.4%)1 (14.3%)1
    (14.3%)
    7 (5.6%)5 (71.4%)1 (14.3%)1
    (14.3%)
    7 (5.7%)1 (50.0%)1 (50.0%)0
    (0.0%)
    2 (1.9%)3 (75.0%)1 (25.0%)0
    (0.0%)
    4 (3.6%)
    Private laboratory10 (47.6%)3 (14.3%)8
    (38.1%)
    21 (16.2%)11 (57.9%)3 (15.8%)5
    (26.3%)
    19 (15.3%)14 (63.6%)3 (13.6%)5
    (22.7%)
    22 (18.0%)20 (87.0%)0 (0.0%)3
    (13.0%)
    23 (21.5%)12 (60.0%)3 (15.0%)5
    (25.0%)
    20 (17.9%)
    Anti-cancer institute4 (44.4%)1 (11.1%)4
    (44.4%)
    9 (6.9%)5 (100.0%)0 (0.0%)0
    (0.0%)
    5 (4.0%)4 (50.0%)2 (25.0%)2
    (25.0%)
    8 (6.6%)7 (70.0%)1 (10.0%)2
    (20.0%)
    10 (9.3%)5 (71.4%)1 (14.3%)1
    (14.3%)
    7 (6.3%)
    Method used                    
    Commercial, non-NGS based kits18 (32.1%)8 (26.7%)26
    (59.1%)
    52 (40.0%)41 (48.2%)7 (38.9%)13
    (61.9%)
    61 (48.4%)34 (40.5%)13 (81.3%)11
    (50.0%)
    58 (47.5%)46 (50.5%)1 (20.0%)5
    (50.0%)
    52 (49.1%)37 (43.0%)6 (50.0%)8
    (57.1%)
    51 (45.5%)
    Non-commercial method23 (41.1%)12 (40.0%)14
    (31.8%)
    49 (37.7%)28 (32.9%)6 (33.3%)5
    (23.8%)
    39 (31.0%)23 (27.4%)2 (12.5%)9
    (40.9%)
    34 (27.9%)18 (19.8%)2 (40.0%)5
    (50.0%)
    25 (23.6%)11 (12.8%)0 (0.0%)0
    (0.0%)
    11 (9.8%)
    NGS6 (10.7%)3 (10.0%)0
    (0.0%)
    9 (6.9%)14 (16.5%)5 (27.8%)3
    (14.3%)
    22 (17.5%)27 (32.1%)1 (6.3%)2
    (9.1%)
    30 (24.6%)27 (29.7%)2 (40.0%)0
    (0.0%)
    29 (27.4%)38 (44.2%)6 (50.0%)6
    (42.9%)
    50 (44.6%)
    NGS by laboratory-developed primers2 (3.6%)2 (6.7%)0
    (0.0%)
    4 (44.4%)3
    (3.5%)
    1 (5.6%)0
    (0.0%)
    4 (18.2%)7 (8.3%)0 (0.0%)0
    (0.0%)
    7 (23.3%)5 (5.5%)0 (0.0%)0
    (0.0%)
    5 (17.2%)3 (3.5%)0 (0.0%)0
    (0.0%)
    3 (6.0%)
    NGS with a commercial panel4 (7.1%)1 (3.3%)0
    (0.0%)
    5 (55.6%)11 (12.9%)4 (22.2%)3
    (14.3%)
    18 (81.8%)20 (23.8%)1 (6.3%)2
    (9.1%)
    23 (76.7%)22 (24.2%)2 (40.0%)0
    (0.0%)
    24 (82.8%)35 (40.7%)6 (50.0%)6
    (42.9%)
    47 (94.0%)
    Combination9 (16.1%)7 (23.3%)4
    (9.1%)
    20 (15.4%)2
    (2.4%)
    0 (0.0%)0
    (0.0%)
    2 (1.6%)0 (0.0%)0 (0.0%)0
    (0.0%)
    0 (0.0%)0 (0.0%)0 (0.0%)0
    (0.0%)
    0 (0.0%)0 (0.0%)0 (0.0%)0
    (0.0%)
    0 (0.0%)
    Performance                    
    Average EQA score (/20)19.018.418.818.818.918.717.118.118.918.817.218,719.019.216.118.619.018.117.818.8
    Success rate91.1%73.3%84.1%110 (84.6%)77.6%66.7%57.1%90 (72.6%)72.6%68.8%50.0%83 (68.0%)65.2%80.0%30.0%66 (61.7%)83.7%91.7%100.0%97 (86.6%)
    Total56 (43.1%)30 (23.1%)44
    (33.8%)
    13085 (68.5%)18 (14.5%)21
    (16.9%)
    12484 (68.9%)16 (13.1%)22
    (18.0%)
    12292 (86.0%)5 (4.7%)10
    (9.3%)
    10786 (76.8%)12 (10.7%)14
    (12.5%)
    112

    ‘Full’ KRAS or NRAS testing includes testing of codons 12, 13 (exon 2), 59, 61 (exon 3), 117 and 146 (exon 4). Testing is defined as ‘incomplete’ if one of the abovementioned codons is not included when testing KRAS or NRAS mutations.

    NGS: Next-generation sequencing.

    Since 2013, an increase was observed in the codons tested in both KRAS and NRAS (Table 1), but full RAS testing (exon 2, 3, and 4 of KRAS and NRAS genes) had not been implemented by all laboratories in 2018. Table 1 shows how full RAS testing was adopted over time for different laboratory settings. In general, the percentage of laboratories who implemented full RAS testing increased between 2013 and 2018 from 43.1% (56/130) to 76.7% (86/112). Of the 112 participants in 2018, 82.9% (29/35) of community hospitals and 80.4% (37/46) of university hospitals included full testing compared with only 71.4% (5/7) of anticancer institutes and 62.5% (15/24) private laboratories/hospitals. It was also demonstrated that between 2013 and 2016 laboratories that applied full RAS testing produced a better EQA absolute score and pass-rate (Table 1). Laboratories which did not perform full RAS testing often did not test for alterations in exon 4 of both genes. In 2017 and 2018 no such variants were included in the EQA scheme. In addition, the scheme of 2018 included the NRAS c.38_40delinsACA p.(Gly13_Val14delinsAspIle) variant. Four laboratories in the ‘full RAS testing group’ were penalized for not correctly detecting this variant by next-generation sequencing, lowering the average score and pass-rate.

    The number of NGS users in the EQA scheme increased between the 2013 and 2018 scheme from 6.9% (9/130) to 44.6% (50/112), in favor of non-NGS noncommercial methods which decreased from 37.7% to 9.8% The number of non-NGS commercial kit users remained stable (40.0% in 2013 vs 45.5% in 2018). Among NGS users, the use of a commercial panel increased over time, in favor of the use of laboratory developed primers (Table 1).

    Across all EQA schemes 5471 tests were analyzable (without technical failures), of which 96.1% (5255/5471) were correct and 3.9% (216/5471) were misclassified. The percentage of misclassifications was the highest for tests performed on samples containing an NRAS variant with 6.0% (33/554) compared with KRAS variants with 4.7% (108/2306), BRAF variants with 4.9% (25/510) or wild-type samples with 2.4% (50/2101). A detailed overview of misclassification results per gene and variants is given in Table 2. The percentage of true false-negative results was 5.4% (152/3370), of which 4.6% (132/2860) for RAS and 3.9% (20/510) for BRAF testing (data not shown).

    Table 2. Number of laboratories testing for KRAS, NRAS and BRAF and misclassification rates per variant among laboratories that participated in the external quality assessment schemes.
    MarkerCodonScheme year20132014201620172018All
      Variant# Laboratories testing codon/# EQA participants (%)# testsMisclassifications (%)# Laboratories testing codon/# EQA participants (%)# testsMisclassifications (%)# Laboratories testing codon/# EQA participants (%)# testsMisclassifications (%)# Laboratories testing codon/# EQA participants (%)# testsMisclassifications (%)# Laboratories testing codon/# EQA participants (%)# testsMisclassifications (%)# testsMisclassifications (%)
    KRAS12c.34G>A p.(Gly12Ser)120/120 (100.0%)  114/115 (99.1%)  114/115 (99.1%)  100/100 (100.0%)  109/109 (100.0%)1110.01110.0
      c.34G>T p.(Gly12Cys) 1313.1            1313.1
      c.35G>A p.(Gly12Asp) 1301.5 1252.4 1072.8 1051.0   4671.9
      c.35G>C p.(Gly12Ala)          1051.9   1051.9
      c.35G>T p.(Gly12Val)       1231.6      1231.6
     13c.38G>A p.(Gly13Asp)119/120 (99.2%)1313.1114/115 (99.1%)1225.7114/115 (99.1%)1220.8100/100 (100.0%)1042.9109/109 (100.0%)1112.75903.1
      c.38_40delinsACA p.(Gly13_Val14delinsAspIle)             1109.11109.1
     59c.176C>A p.(Ala59Glu)20/120 (16.7%)  95/115 (82.6%)  101/115 (87.8%)  97/100 (97.0%)10211.8102/109 (93.6%)  10211.8
     61c.182A>T p.(Gln61Leu)105/120 (87.5%)  114/115 (99.1%)244.2111/115 (96.5%)  99/100 (99.0%)  108/109 (99.1%)  244.2
      c.183A>C p.(Gln61His)    577.1         577.1
      c.183A>T p.(Gln61His)    4413.6       1120.91567.3
     117c.35G>A p.(Gly12Asp)82/120 (68.3%)  105/115 (91.3%)  110/115 (95.6%)1566.799/100 (99.0%)  106/109 (97.2%)  1566.7
      + c.351A>T p.(Lys117Asn)                 
     146c.436G>A p.(Ala146Thr)84/120 (70.0%)773.9108/115 (93.9%)12010.0111/115 (96.5%)11811.999/100 (99.0%)  106/109 (97.2%)  3159.2
     AllSubtotal KRAS 4692.8 4926.7 4856.2 4164.3 4443.223064.7
    NRAS1294/96 (97.9%)  112/112 (100.0%)  112/112 (100.0%)  100/100 (100.0%)  109/109 (100.0%)    
     1338/96 (39.6%)  110/112 (98.0%)  110/112 (98.2%)  99/100 (99.0%)  108/109 (99.1%)    
     5923/96 (24.0%)  93/112 (83.0%)  100/112 (89.2%)  97/100 (97.0%)  103/109 (94.5%)    
     61c.181C>A p.(Gln61Lys)96/96 (100.0%)9713.4112/112 (100.0%)156.7112/112 (100.0%)728.3100/100 (100.0%)  109/109 (100.0%)1121.82967.4
      c.182A>G p.(Gln61Arg)    1084.6 150.0      1234.1
      c.182A>T p.(Gln61Leu)       316.5 1043.8   1354.4
     11734/96 (35.4%)  88/112 (78.6%)  96/112 (85.7%)  95/100 (95.0%)  101/109 (92.6%)    
     14641/96 (42.7%)  100/112 (89.3%)  109/112 (97.3%)  99/100 (99.0%)  105/109 (96.3%)    
     AllSubtotal NRAS 9713.4 1234.9 1186.8 1043.8 1121.85546.0
    BRAF600c.1799T>A p.(Val600Glu)1035.81047.71065.7914.41060.95104.9
    KRAS, NRAS, BRAFAllWild-type 5072.0 4765.1 3652.7 3151.6 4380.221012.4
    Total   11763.6 11955.9 10745.1 9263.3 11001.654713.9

    Misclassifications included false-positive results (identification of a variant in a wild-type case, or an additional variant besides the correct variant), false-negative results (variant in the sample unidentified) and wrong mutations that affected the mutation rate (incorrect variant in the same or another gene of interest in a sample containing a variant) variant in another gene reported resulting in an FN or FP results for the respective gene).

    EQA: External quality assessment; FN: False-negative; FP: False-positive; N: Number of tests.

    The percentage of misclassifications observed during the EQA scheme increased initially from 3.6% (42/1176) in 2013 toward 5.9% (71/1195) in 2014. Thereafter, misclassification rates decreased toward 1.6% (18/1100) in 2018 (Figure 1A). This coincided with an initial increase in FN results from 4.2% (28/669) in 2013 to 5.8% (42/719) in 2014 and 5.9% (42/709) in 2015. Thereafter, FNs decreased also to 2.6% (16/662) in 2018 (Figure 1B).

    Figure 1. Overview of rates (in %) of misclassification (A), true false-negatives (B) and technical failures (C) on test level in the different external quality assessment scheme years.

    (A) Misclassification rates include the total of false-positive, false-negative and incorrect mutations affecting the mutation rate on the total of tests with an outcome, with exclusion of the technical failures. If the gene of interest was not included in the test methodology this was not counted as a misclassification. (B) The true false-negative rates represent only the number of false-negative results on the total of tests with an outcome, with exclusion of the technical failures. No false-negative rate is available for the WT cases as these samples did not include a variant. (C) Technical failures are the number of tests for which no outcome could be given due to a test failure, on the total number of tests performed. All rates are given in percentages for the total cases distributed in the schemes, cases with a KRAS, NRAS or BRAF variant and WT cases, respectively. The number of tests is represented below the respective category. In 2015, no EQA scheme was organized.

    EQA: External quality assessment; WT: Wild-type.

    A decrease in misclassifications was also observed after repeated participation to EQA scheme rounds. Namely, for laboratories who participated only once (irrespective of the scheme year), 5.4% of tests (116/2156) were incorrectly analyzed compared with 0.2% of tests (1/422) for laboratories in 2018, who took part in all five successive scheme years (Figure 2A). The fraction of false-negative results declined simultaneously from 5.9% (75/1277) to 0.4% (1/254) between a first and fifth participation, respectively (Figure 2B).

    Figure 2. Overview of rates (in %) of misclassification (A), true false-negatives (B) and technical failures (C) on test level related to the number of successive external quality assessment schemes a laboratory took part in.

    (A) Misclassification rates include the total of false-positive, false-negative and incorrect mutations affecting the mutation rate on the total of tests with an outcome, with exclusion of the technical failures. If the gene of interest was not included in the test methodology this was not counted as a misclassification. (B) The true false-negative rates represent only the number of false-negative results on the total of tests with an outcome, with exclusion of the technical failures. No false-negative rate is available for the WT cases as these samples did not include a variant. (C) Technical failures are the number of tests for which no outcome could be given due to a test failure, on the total number of tests performed. All rates are given in percentages for the total cases distributed in the schemes, cases with a KRAS, NRAS or BRAF variant and WT cases, respectively. The number of tests is represented below the respective category. In 2015, no EQA scheme was organized. Numbers 1, 2, 3, 4, 5 represents the number of successive EQA schemes a laboratory participated to.

    EQA: External quality assessment; WT: Wild-type.

    A wide range of test methodologies was used by the participants for KRAS, NRAS and BRAF analysis with a variety of misclassification and failure percentages (Table 3). All genes combined, more misclassifications were observed in tests performed by non-NGS based commercial test kits with 4.3% (186/4372) compared with non-NGS/noncommercial (laboratory-developed) tests with 2.6%, (69/2693) and NGS-based techniques with 1.3% (29/2207).

    Table 3. Overview of misclassification rates and technical failure rates (in %) on test level related to the used testing methodologies.
    MarkerKRASNRASBRAFTotal
    Test methodologyNTF (%)MC (%)NTF (%)MC (%)NTF (%)MC (%)NTF (%)MC (%)
    Commercial kit22121.34.711391.34.310211.83.343721.44.3
    Therascreen KRAS Pyro Kit (Qiagen DxS)4260.74.0      4260.74.0
    Cobas® KRAS Mutation Test (Roche)3811.33.9      3811.33.9
    KRAS mutation analysis kit (EntroGen)2462.45.7      2462.45.7
    Idylla KRAS Mutation Test (Biocartis)2181.81.4      2181.81.4
    Therascreen KRAS RGQ PCR Kit (Qiagen DxS)1542.67.1      1542.67.1
    Myriapod Colon Status (Diatech Pharmacogenetics)9801640058002200.00.5
    KRAS/BRAF mutation analysis kit (EntroGen)951.15.3   392.65.11341.55.2
    KRAS StripAssay (ViennaLab)881.18.0      881.18.0
    KRAS XL StripAssay (ViennaLab)831.24.8      831.24.8
    KRAS 19 Mutations Detection Kit (Amoy Diagnostics)810.07.4      810.07.4
    Anti EGFR MoAb response /(KRAS status) (Diatech Pharmacogenetics)521.91.9      521.91.9
    Therascreen NRAS Pyro Kit (Qiagen Dxs)   2703.05.2   2703.05.2
    NRAS Mutation Detection Kit (Entrogen)   1640.67.9   1640.67.9
    Idylla NRAS-BRAF assay (Biocartis)   1103.61.81134.40.92234.01.3
    Therascreen RAS extension Pyro Kit (Qiagen)   870.00   870.00.0
    BRAF/NRAS Mutation Test (Roche)   700.00590.03.41290.01.6
    NRAS XL StripAssay (ViennaLab)   630.03.2   630.03.2
    NRAS Mutation Detection Kit (Amoy Diagnostics)   530.05.7   530.05.7
    KRAS/NRAS Lightmix Kit (Tib Molbiol)   510.017.6   510.017.6
    Therascreen BRAF Pyro Kit (Qiagen DxS)      1953.62.61953.62.6
    Cobas 4800 BRAF V600 Mutation Test (Roche)      1840.06.01840.06.0
    BRAF Mutation Analysis Kit (EntroGen)      1150.91.71150.91.7
    BRAF StripAssay (ViennaLab)      610.01.6610.01.6
    Other (38)2901.06.62071.02.91972.05.16941.76.7
    Noncommercial method12331.73.17902.02.46701.51.826931.72.6
    Dideoxy sequencing8811.22.86032.02.33931.81.318771.62.3
    Pyrosequencing1274.73.1725.61.4890.01.12883.52.1
    Allelic driscrimination (with Taqman probes)722.82.8   541.91.91262.42.4
    Other (15)1531.34.61150.03.51341.53.74021.02.9
    Next-generation sequencing9952.21.66192.61.15932.91.022072.51.3
    Ion AmpliSeq Colon and Lung Cancer Panel (Life Technologies)2381.70.01420.70.01420.70.05221.10.0
    In-house primer set1241.64.8732.74.1521.95.82492.04.8
    Ion Ampliseq Custom panel – Regions selected by the laboratory (Life Technologies)1213.30.8734.11.4736.80.02674.50.7
    TruSight Tumor OncoPanel/TruSight Tumor 15 (Illumina)9201.1560.00.0560.00.02040.00.5
    Tumor Hotspot MASTR Plus (Multiplicom)702.91.4434.72.3434.70.01563.81.3
    Ion AmpliSeq Cancer Hotspot Panel v2 (Life Technologies)621.60.0382.62.6382.60.01382.20.7
    TruSeq Amplicon Cancer Panel (Illumina)618.24.93713.52.73713.55.413511.14.4
    Actionable Mutations panel (Qiagen)551.81.8342.90342.90.01232.40.8
    Other (13)1721.71.71230.80.01180.80.84131.21.4
    Grand Total44401.63.525481.82.922842.02.392721.83.1

    The total number of tests is given over all EQA scheme years (2013–2018). Only data for which both, method information and analysis scores were available, are included. Only methods for which at least 50 tests were performed were included.

    †Other methods are represented in Supplementary Table 3. Misclassification rates are calculated on the number of tests for that respective gene, summed with the total of tests on wild-type samples by the same methodology. Misclassifications included: false-positive results (identification of a variant in a wild-type case, or an additional variant besides the correct variant), false-negative results (variant in the sample unidentified) and wrong mutations that affected the mutation rate (incorrect variant in another gene reported resulting in an FN or FP results for the respective gene).

    EQA: External quality assessment; FN: False-negative; FP: False-positive; N: Number of tests; MC: Misclassification; TF: Technical failure.

    To evaluate the influence of a change in test method, 25.5% (336/1320) of data points were excluded, as these were new laboratories and no method information was available from the previous EQA scheme. For the remaining 74.5% (984/1320) of data, a switch in testing methodology was observed in 35.5% (349/984) individual laboratory participations (data not shown). There was no observed difference in errors or technical failure rates between laboratories who changed and those who did not change test methodologies (data not shown).

    In total, 1.6% (91/5562) technical failures were observed, which were more frequent for wild-type samples with 2.2% (48/2149) compared with cases containing KRAS (1.1%, 25/2331), NRAS (1.6%, 9/563) or BRAF (1.7%, 9/519) variants (data not shown) and for NGS (2.5%, 55/2207) compared with the other testing strategies (1.4%, 62/4372 and 1.7%, 47/2693 for commercial and noncommercial tests, respectively; Table 3). Technical failures diminished between 2013 and 2018 from 2.9% (35/1211) to 1.3% (15/1115 tests; Figure 1C) and for laboratories who participated to EQA more frequently (Figure 2C). Namely, participants on average reported 2.3% (50/2206) of technical failures during their first participation, compared with 0.3% (2/607) and 1.4% (6/428) during their fourth and fifth participation, respectively.

    Pathology data collection via a survey

    In addition to data from the EQA database, the 200 laboratories participating to the EQA schemes between 2013 and 2017 were invited to provide additional information via a survey. 95 laboratories initiated the online link of which 33 laboratories from 28 countries provided the information. Of those 33 laboratories, 21 laboratories provided information for the full study period, three (2013–2018) provided data for 2014 to 2017, four provided information for 2015 to 2017 and two provided data for 2016 to 2017.

    The majority of participating laboratories (78.8%; 26/33) reported to test RAS on request by the oncologist only, whereas seven laboratories (21.2%; 7/33) reported routinely testing all CRC/mCRC patients (Q3 from survey, see Supplementary Data 1). For the remaining laboratories, testing was performed on request by either the oncologist or the pathologist (n = 2), or by other hospitals (n = 1). Between 2013 and 2017, the 33 laboratories provided information on 27,325 samples from mCRC patients, which were tested for RAS mutations. There was a small increase on the average numbers of mCRC samples tested from 246.5 in 2013 to 271.1 in 2018 (Table 4). A total of 18 (54.5%) laboratories reported to have a ‘notable’ increase in the number of samples tested during the period of 2013 to 2017, whereas three reported a ‘notable’ decrease (Q5 from survey, see Supplementary Data 1). The most common reasons the laboratories provided for ’notable’ testing increases were ‘more routine testing occurring’ with 50.0% (9/18) and ‘larger catchment area’ with 22.2% (4/18) (Q5a from survey, see Supplementary Data 1). A total of 17% (3/18) of laboratories thought both an increase in routine testing and testing a larger catchment area resulted in the increase. A total of 11% (2/18) of laboratories reported other reasons for this increase. The overall RAS mutation rate in the tested samples was 45.2% (12,341/27,325, 95% CI: [44.6%–45.8%]). There was no consistent change between 2013 and 2017 with the annual estimates varying between 42.6% (998/2343; 95% CI: [30]) in 2013 and 47.0% (3586/7636; 95% CI: [45.8%–48.1%]) in 2016 (Table 4).

    Table 4. Number of RAS samples tested and average number of samples tested per survey respondent by year related to RAS mutation rate over time.
    Number of samples testedYear
     Total20132014201520162017
    Number of unique labs311924293131
    Total number of samples tested27,32523435638750576364203
    Annual average samples per laboratory (95% CI)203.9 (141.1–266.8)123.3 (85.5–161.1)235.0 (155.5–314.3)258.8 (185.1–332.5)246.3 (171.8–320.9)135.6 (101.7–169.4)
    Projected annual samples per laboratory (95% CI)252.8 (184.2– 321.3)246.5 (170.9– 322.1)235.0 (155.5– 314.3)258.8 (185.1–332.5)246.3 (171.8– 320.9)271.1 (203.5– 338.9)
    Mutation rates
    Mutant12,3419982578326835861911
    Wild-type14,0321.2602810400838082146
    Unknown7075720217918881
    % Mutant (95% CI)45.2 (44.6–45.8)42.6 (40.1–44.6)45.7 (44.4–47.0)43.5 (42.4–44.7)47.0 (45.8–48.1)45.5 (44.0–47.0)

    †Data available for 6 months only.

    ‡For 2013 and 2017 the sample numbers were doubled to produce comparable estimates.

    Discussion

    The EMA label for anti-EGFR mAbs was extended to full RAS testing in 2013. This study aimed to provide an overview of RAS biomarker testing in Europe, in relation to mutation rates and testing practices, based on data from the ESP Colon EQA program and a European-wide survey. The results demonstrated an overall RAS mutation rate in the tested samples of 45.2% (12,341/27,325, 95% CI: [44.6–45.8%]), which is in line with other observational studies of RAS mutations in Europe [24,31]. Although this is substantially lower than the 55.9% (95% CI: [53.9–57.9%]) reported in some clinical trials [32]. A possible explanation could be that in the study by Peeters et al. the majority of patients were tested in the context of second-line therapy or best supportive care, whereas this is unknown for our current study. Moreover, as RAS status was determined for 1014 patients (compared with more than 27,000 cases in our study), the chance of obtaining a RAS mutation positive case would be higher. The inclusion of this large dataset on tested samples in this study, allows a solid estimation of the mutation rate, as even the rarest variants in these genes have been reported at a frequency of ≥1% [33].

    There was no consistent change in RAS mutation rate over time. One would have expected that if laboratories are now testing for more alterations in RAS and are improving their experience and performance over time, the RAS mutation rate would have also increased. The results of this study clearly illustrate that this is not the case. A plausible explanation could be that alterations in exons 3 and 4 of KRAS and NRAS genes are rather rare and therefore have little impact on the total mutation rate. Given that information on specific rare variants rates is missing in the early years, further investigation may be required to evaluate mutation rates of less frequent variants in the different RAS codons to verify if mutations in specific codons were missed specifically in the first years. In addition, information on mutation rates before the change of the drug labels in 2013 might provide a good estimate of the mutations that were missed at the time that exon 4 mutations were not yet included. However, retrieving this information might be difficult and therefore, retesting of leftover samples may be necessary.

    Most laboratories that participated in the EQA programs between 2013 and 2018 have implemented full RAS testing over time, however, KRAS and NRAS codons 59, 117 and 146 remain untested by a group of laboratories. Especially in the early years after 2013, laboratories had separate kits for KRAS exon 2 testing and secondary for the other KRAS exons and NRAS. If the patient had a KRAS exon 2 mutation, laboratories could save money by not performing further tests for the other exons. For NRAS for instance, the two most widely used kits for NRAS analysis Therascreen NRAS Pyro Kit (Qiagen DxS) and Cobas®NRAS Mutation Test (Roche) only include codons 12, 13 and 61 of the NRAS gene (Supplementary Table 2). Eight out of nine laboratories, which are not yet offering full RAS testing anno 2018, declared to use a non-NGS based commercial kit, which does not include all codons. The other laboratory used a NGS-based commercial kit. Important alterations could be present in the codons for which testing is not fully implemented yet, such as the NRAS c.181C>A p.(Gln61Lys) variant, which accounts for 27.2% of NRAS mutations (COSMIC).

    The laboratories offering full RAS testing right after the label change were typically university hospitals, with on average high scores during EQA participation (≥18/20 points). In a previous study from Tack et al., it was observed that laboratories that also perform testing in a research setting (as opposed to only in a clinical setting), and laboratories which are accredited, had a better performance for NRAS testing in the first years after the label change (2013–2014), with less analysis errors being were made [34]. In the ESP Colon EQA schemes, no points were deduced when a laboratory did not test for rare variants in exons 3 and 4. Even with this milder scoring system, laboratories with incomplete RAS testing scored worse than those with full RAS testing. From a clinical point of view, it is time for EQA programs for being stricter with regard to rare variants, and demand testing for all clinically relevant mutations. Participants are advised to apply a testing method covering all relevant mutations, and to review relevant literature as more mutations might become clinically relevant in the future.

    In this study, an overall percentage of true FN RAS results of 4.6% (132/2860, all scheme years) was observed, which might have serious implications for patient safety by altering the decision on targeted therapy administration. (Figure 1B). It must be noted that EQA performance is only an indication of routine practice. However, recently it was shown that laboratories performing well in EQA also perform well in routine testing, stressing the importance of these results for daily practice [35].

    The observed FN and overall misclassification rates initially increased over time and where a laboratory participated in more EQA schemes, resulted in an improvement toward 2018. This initial increase could be explained by the more stringent testing criteria for full RAS testing, or by the lack of laboratory experience with testing multiple markers or participating to the EQA scheme. The following improvement coincides with the shift toward full RAS testing and the more sensitive techniques. Higher FN or misclassification rates during the first year (2013) or first participation, were especially observed for NRAS, presumably because laboratories already had more prior experience with KRAS testing compared with NRAS generally. Also, more misclassifications occurred for RAS variants in codons 59, 61, 117 and 146 compared with codons 12 and 13 (Table 2), suggesting that although routinely tested for several years, laboratories have still not fully adopted less frequent mutations. In addition, more misclassifications were observed in 2018 for a complex KRAS deletion–insertion compared with other point mutations, of which 90.0% (9/10) included FN results (wild-type reported) and in 10.0% (1/10) a KRAS codon 146 variant was reported instead. Increased error rates for complex mutations have been described in non-small-cell lung cancer as well. Even though BRAF is not yet approved for predictive testing, performance for this biomarker was similarly compared with those observed for RAS analysis. While one would expect more misclassifications for laboratory-developed noncommercial techniques, due to their limited sensitivity, our analysis revealed that those methods performed slightly better when compared with commercial test kits (Table 3). This may be explained by the fact that commercial test kits often do not include all necessary codons for testing, as described earlier, and the danger of FP results with high-sensitive mutation specific analyses.

    On the contrary, more technical failures were made by NGS compared with other test methodologies. Since all distributed samples contained at least 30% neoplastic cells, which is above the minimum requirement of all test methods used, the effect of the neoplastic cell percentage estimates prior to DNA extraction was not included to calculate the misclassification or technical failure rates in this study. Nevertheless, this estimation has previously been reported to be highly variable, which might be at the base of the higher failure rate for NGS as an accurate neoplastic cell content estimation helps to set a minimal read depth, because samples with a low percentage of tumor cells require higher coverages [36]. For the 0.1% (91/5562) technical failures the main reasons were a too low quality of the extracted DNA (34.1%, 31/91), DNA concentration too low (12.1%, 11/91) or a too low percentage of neoplastic cells in the sample (9.9%, 9/91). Survey results indicated that the laboratories are indeed taking the necessary quality control actions after DNA extraction prior to proceeding with the analysis itself, with the majority of respondents quantifying the DNA concentration by a spectrophotometer (54.4%, 18/33) or another fluorescence method (39.4%, 13/33) (Supplementary Data 1, Q4). The fact that laboratories report technical failures more frequently in wild-type cases could be explained by their insecurity of interpretation of the test result. Namely, in case of too low DNA concentration, a wild-type result does not exclude the presence of a mutation which was not detected, while detection of a variant can be interpreted with more certainty.

    The study provided average number of samples tested per laboratory per year, which determined a limited increase of the average number of samples tested between 2013 and 2017. These estimates should be interpreted with caution as they do not strictly correspond to the proportion samples tested for RAS. This is because there is no fixed denominator as the patient catchment area for each laboratory can vary by year. Indeed, out of the 18 laboratories that reported a ‘notable’ increase in samples tested, 50.0% (9/18) was reportedly due to a larger patient catchment area whereas 22.0% (4/18) was due to more samples being tested (Q5 from survey, Supplementary Data 1). The latter is reflected by the increasing incidence rate of mCRC in Europe [37]. Although here the average number of tested cases per year is presented across all respondents, previous research has indicated that testing a larger number of samples per year can serve as a predictive factor for a better performance during EQA [34].

    To the best of our knowledge, with approximately 27000 mCRC samples tested for RAS, this study provides the most robust estimates of real-world RAS mutation rates to-date. Moreover, it is the first time that mutation prevalences were linked to laboratory practices to further understand their impact. The results showed no trend of RAS mutation rate over time despite having a clear shift to more sensitive tests and increased quality of the testing methods. The overall landscape of RAS testing in Europe appears to be positive with highly sensitive and specific tests. Since testing techniques have no impact, this mutation rate can now be used for a future follow-up of the mutation rate in population of patients with mCRC. However, as there still appears to be a small amount of misclassification it is important that laboratories continue to improve their testing methods and should all implement full RAS testing. Although NGS is becoming the gold standard for full testing, technical parameters (DNA quality and percentage of neoplastic cells) must be considered to prevent technical failures. The current study only presented data from a laboratory point of view. It could be further studied whether clinicians correctly interpret the test results and translate it to the right choice for therapy.

    Future perspective

    It is suspected that there will be further studies that will confirm the RAS mutation rate on a large scale. Although new mutations with an effect on the response to anti-EGFR therapy are becoming known, the impact on the RAS mutation rate will be limited as these are rather ‘rare mutations’.

    The performance of the laboratories is currently high, with 96.4% of the samples correctly analyzed, yet it is expected that the laboratories will strive toward 100% accuracy. This is important given the effect the test result has on the response to anti-EGFR therapy. A first step toward this increased performance will be the further expansion of the number of codons tested and also the implementation of broad-testing and sensitive methods. A possible shift to testing on liquid biopsy samples is also expected, for which even more sensitive methods are needed than for tissue samples. It is important that when implementing these novelties, standard procedures are used for test validation and -verification and internal quality control. Earlier results of EQA schemes showed that this ‘quality-way of thinking’ is already known in Europe, but that there is still room for improvement.

    Summary points
    • Two monoclonal antibody therapies against EGFR are approved (panitumumab and cetuximab) for patients with a RAS wild-type status.

    • For confirming a RAS wild-type status, the following codons in both the KRAS and NRAS gene should be tested: 12, 13, 59, 61, 117, 146.

    • Between 2013 and 2017, 33 laboratories provided information on 27,325 samples from mCRC patients that were tested for RAS. The overall RAS mutation rate in the tested samples was 45.2% (12,341/27,325, 95% CI: [44.6%, 45.8%]), without a variation over time.

    • Laboratory performance was studied based on data from 239 European laboratories which participated to a European external quality assessment (EQA) scheme for metastatic colorectal cancer (data from 2013 to 2018).

    • The mutation status was correctly analyzed in 5255/5471 (96.1%) RAS and BRAF tests. The performance increased between 2013 and 2018.

    • 132/2860 (4.6%) RAS tests included FN results.

    • The percentage of misclassifications was the highest for tests performed on samples containing an NRAS variant with 6.0% (33/554) compared with KRAS variants with 4.7% (108/2306), BRAF variants with 4.9% (25/510) or wild-type samples with 2.4% (50/2101).

    • The percentage of laboratories who implemented full RAS testing increased between 2013 and 2018 from 43.1% (56/130) to 76.7% (86/112). In 2018, 23.2% (26/112) does not test all required codons for anti-EGFR therapy.

    • The number of laboratories using next-generation sequencing in the EQA scheme increased between the 2013 and 2018 scheme from 6.9% (9/130) to 44.6% (50/112).

    • All genes combined, more misclassifications were observed in tests performed by non-NGS based commercial test kits (4.3%; 186/4372) compared with NGS-based techniques with (1.3%).

    Supplementary data

    To view the supplementary data that accompany this paper please visit the journal website at: www.futuremedicine.com/doi/suppl/10.2217/crc-2019-0013

    Author contributions

    EQA analysis: C Keppens and EMC Dequeker conceived and designed the study set-up. K Dufraing, C Keppens, V Tack and EMC Dequeker were responsible for collection of the EQA scheme data according to ISO17043. JH van Krieken provided medical expertise during the EQA schemes. M Ligtenberg was involved in technical expertise and sample validation during the EQA schemes. K Dufraing, C Keppens, V Tack, JH van Krieken, M Ligtenberg, N Normanno, S Vander Borght, LI Kroeze, L Tembuyser assessed the EQA results. K Dufraing, C Keppens and EMC Dequeker analyzed and interpreted the EQA scheme data.

    Pathology survey: V Tack, AG Siebers, G Kafatos, S Dube, G Demonty, K Lowe, JH van Krieken and EMC Dequeker conceived and designed the study set-up. V Tack, S Dube, G Kafatos and EMC Dequeker were responsible for creation, sending and result collection of the survey. K Dufraing, C Keppens, V Tack, S Dube analyzed and interpreted the collected data.

    K Dufraing, C Keppens and EMC Dequeker were responsible for writing of the manuscript. All authors critically revised the manuscript for important intellectual content.

    Acknowledgments

    This project would not have been possible without the support of the following people: All laboratories that participated to the ESP Colon EQA schemes and/or completed the survey, for their contribution in providing of the data. The ESP QA Foundation, for the administrative support during the schemes. The regional scheme organizers for validation and preparation of the 2016 EQA samples: B Bisig, B Savaş, E Rouleau, G Höfler, G De Hertogh, G Troncone, JC Machado, K De Stricker, R Willems. Our colleagues at the Biomedical Quality Assurance Research Unit for coordination and administrative support during the schemes.

    Financial & competing interests disclosure

    This study was funded by a research grant from Amgen Inc., Thousand Oaks, CA 91320, USA (grant number not applicable). G Kafatos, G Demonty and K Lowe are employees and hold shares of Amgen, Sabada Dube is a former employee of Amgen. JH Van Krieken received research grant funding from Amgen. The authors have no other relevant affiliations or financial involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript apart from those disclosed.

    No writing assistance was utilized in the production of this manuscript.

    Ethical conduct of research

    The authors state that they have obtained appropriate institutional review board approval or have followed the principles outlined in the Declaration of Helsinki for all human or animal experimental investigations. In addition, for investigations involving human subjects, informed consent has been obtained from the participants involved.

    Open access

    This work is licensed under the Attribution-NonCommercial-NoDerivatives 4.0 Unported License. To view a copy of this license, visit http://creativecommons.org/licenses/by-nc-nd/4.0/

    Papers of special note have been highlighted as: • of interest; •• of considerable interest

    References

    • 1. Ferlay J , Soerjomataram I , Dikshit R et al. Cancer incidence and mortality worldwide: sources, methods and major patterns in GLOBOCAN 2012. Int. J. Cancer 136(5), E359–E386 (2015).
    • 2. Ferlay J , Steliarova-Foucher E , Lortet-Tieulent J et al. Cancer incidence and mortality patterns in Europe: estimates for 40 countries in 2012. Eur. J. Cancer 49(6), 1374–1403 (2013). •• Gives an overview of the incidence and mortality estimates for different cancer types.
    • 3. Bolocan A , Ion D , Ciocan DN , Paduraru DN . Prognostic and predictive factors in colorectal cancer. Chirurgia (Bucur) 107(5), 555–563 (2012).
    • 4. Kurkjian C , Kummar S . Advances in the treatment of metastatic colorectal cancer. Am. J. Ther. 16(5), 412–420 (2009).
    • 5. van der Geest LG , Koopman M , Verhoef C , Elferink MA , de Wilt JH . Nationwide trends in incidence, treatment and survival of colorectal cancer patients with synchronous metastases. Clin. Exp. Metastasis 32(5), 457–465 (2015).
    • 6. Van der Pool A , Damhuis R , Ijzermans J et al. Trends in incidence, treatment and survival of patients with stage IV colorectal cancer: a population‐based series. Colorectal Dis. 14(1), 56–61 (2012).
    • 7. Mitchell EP . Targeted therapy for metastatic colorectal cancer: role of aflibercept. Clin. Colorectal Cancer 12(2), 73–85 (2013).
    • 8. Van Cutsem E , Kohne CH , Lang I et al. Cetuximab plus irinotecan, fluorouracil, and leucovorin as first-line treatment for metastatic colorectal cancer: updated analysis of overall survival according to tumor KRAS and BRAF mutation status. J. Clin. Oncol. 29(15), 2011–2019 (2011).
    • 9. Douillard JY , Oliner KS , Siena S et al. Panitumumab-FOLFOX4 treatment and RAS mutations in colorectal cancer. N. Engl. J. Med. 369(11), 1023–1034 (2013).
    • 10. Peeters M , Price TJ , Cervantes A et al. Randomized Phase III study of panitumumab with fluorouracil, leucovorin, and irinotecan (FOLFIRI) compared with FOLFIRI alone as second-line treatment in patients with metastatic colorectal cancer. J. Clin. Oncol. 28(31), 4706–4713 (2010).
    • 11. Giantonio BJ , Catalano PJ , Meropol NJ et al. Bevacizumab in combination with oxaliplatin, fluorouracil, and leucovorin (FOLFOX4) for previously treated metastatic colorectal cancer: results from the Eastern Cooperative Oncology Group Study E3200. J. Clin. Oncol. 25(12), 1539–1544 (2007).
    • 12. Kim TW , Elme A , Kusic Z et al. A Phase 3 trial evaluating panitumumab plus best supportive care vs best supportive care in chemorefractory wild-type KRAS or RAS metastatic colorectal cancer. Br. J. Cancer 115(10), 1206 (2016).
    • 13. Erbitux. Erbitux® (cetuximab) [package insert]. Bristol-Myers Squibb Company. ImClone Systems Incorporated and Princeton, NJ, NY, USA (2006).
    • 14. Vectibix. Vectibix® (panitumumab) [Package insert]. Amgen Inc., CA, USA (2008).
    • 15. Benvenuti S , Sartore-Bianchi A , Di Nicolantonio F et al. Oncogenic activation of the RAS/RAF signaling pathway impairs the response of metastatic colorectal cancers to anti-epidermal growth factor receptor antibody therapies. Cancer Res. 67(6), 2643–2648 (2007).
    • 16. Di Fiore F , Blanchard F , Charbonnier F et al. Clinical relevance of KRAS mutation detection in metastatic colorectal cancer treated by Cetuximab plus chemotherapy. Br. J. Cancer 96(8), 1166–1169 (2007). •• Cnfirms that the NRAS mutation is predictive for a lack of response to cetuximab in mCRC and that highly sensitive testing methods are needed.
    • 17. De Roock W , Claes B , Bernasconi D et al. Effects of KRAS, BRAF, NRAS, and PIK3CA mutations on the efficacy of cetuximab plus chemotherapy in chemotherapy-refractory metastatic colorectal cancer: a retrospective consortium analysis. Lancet Oncol. 11(8), 753–762 (2010).
    • 18. Freeman DJ , Juan T , Reiner M et al. Association of K-ras mutational status and clinical outcomes in patients with metastatic colorectal cancer receiving panitumumab alone. Clin. Colorectal Cancer 7(3), 184–190 (2008). •• Confirms that metastatic colorectal cancer patients with a NRAS mutation respond less to panitumumab. Also a NRAS mutation rate of 38.7% is given for the samples analyzed.
    • 19. Lievre A , Bachet JB , Boige V et al. KRAS mutations as an independent prognostic factor in patients with advanced colorectal cancer treated with cetuximab . J. Clin. Oncol. 26(3), 374–379 (2008).
    • 20. De Roock W , Piessevaux H , De Schutter J et al. KRAS wild-type state predicts survival and is associated to early radiological response in metastatic colorectal cancer treated with cetuximab. Ann. Oncol. 19(3), 508–515 (2008).
    • 21. Karapetis CS , Khambata-Ford S , Jonker DJ et al. K-ras mutations and benefit from cetuximab in advanced colorectal cancer . N. Engl. J. Med. 359(17), 1757–1765 (2008).
    • 22. Taieb J , Balogoun R , Le Malicot K et al. Adjuvant FOLFOX +/- cetuximab in full RAS and BRAF wildtype stage III colon cancer patients. Ann. Oncol. 28(4), 824–830 (2017).
    • 23. Kaczirek K , Ciuleanu TE , Vrbanec D et al. FOLFOX4 plus cetuximab for patients with previously untreated metastatic colorectal cancer according to tumor RAS and BRAF mutation status: updated analysis of the CECOG/CORE 1.2.002 study. Clin. Colorectal Cancer 14(2), 91–98 (2015).
    • 24. Kafatos G , Niepel D , Lowe K et al. RAS mutation prevalence among patients with metastatic colorectal cancer: a meta-analysis of real-world data . Biomark. Med. 11(9), 751–760 (2017).
    • 25. Tejpar S , Bertagnolli M , Bosman F et al. Prognostic and predictive biomarkers in resected colon cancer: current status and future perspectives for integrating genomics into biomarker discovery. Oncologist 15(4), 390–404 (2010).
    • 26. Mao C , Liao RY , Qiu LX , Wang XW , Ding H , Chen Q . BRAF V600E mutation and resistance to anti-EGFR monoclonal antibodies in patients with metastatic colorectal cancer: a meta-analysis . Mol. Biol. Rep. 38(4), 2219–2223 (2011).
    • 27. van Krieken JH , Normanno N , Blackhall F et al. Guideline on the requirements of external quality assessment programs in molecular pathology. Virchows Arch. 462(1), 27–37 (2013).
    • 28. International Organization for Standardization. ISO 17043:2010 Medical laboratories – conformity assessment – general requirements for proficiency testing. 2010 ISO Geneva, Switzerland.
    • 29. Tack V , Ligtenberg MJ , Tembuyser L et al. External quality assessment unravels interlaboratory differences in quality of RAS testing for anti-EGFR therapy in colorectal cancer. Oncologist 20(3), 257–262 (2015).
    • 30. Fassunke J , Ihle MA , Lenze D et al. EGFR T790M mutation testing of non-small cell lung cancer tissue and blood samples artificially spiked with circulating cell-free tumor DNA: results of a round robin trial. Virchows Arch. 471(4), 509–520 (2017).
    • 31. Boleij A , Tack V , Taylor A et al. RAS testing practices and RAS mutation prevalence among patients with metastatic colorectal cancer: results from a Europe-wide survey of pathology centres. BMC Cancer 16(1), 825 (2016).
    • 32. Peeters M , Oliner KS , Price TJ et al. Analysis of KRAS/NRAS Mutations in a Phase III study of panitumumab with FOLFIRI compared with FOLFIRI alone as second-line treatment for metastatic Colorectal Cancer. Clin. Cancer Res. 21(24), 5469–5479 (2015). •• Shows that patients with RAS mutations were unlikely to benefit from panitumumab. The RAS mutation rate observed in this study was 58%.
    • 33. Lièvre A . Des mutations de KRAS aux mutations de RAS: vers une meilleure définition de la réponse aux anticorps anti-EGFR dans le cancer colorectal métastatique. Oncologie 16, 120–128 (2014).
    • 34. Tack V , Schuuring E , Keppens C et al. Accreditation, setting and experience as indicators to assure quality in oncology biomarker testing laboratories. Br. J. Cancer 119(5), 605–614 (2018). • Explains which laboratory characteristics are correlated with better performance in the field of predictive biomarker testing in oncology.
    • 35. Tack V , Ligtenberg MJL , Siebers AG et al. RAS testing for colorectal cancer patients is reliable in European laboratories that pass external quality assessment. Virchows Arch. 472(5), 717–725 (2018).
    • 36. Dufraing K , De Hertogh G , Tack V , Keppens C , Dequeker EMC , van Krieken JH . External quality assessment identifies training needs to determine the neoplastic cell content for biomarker testing. J. Mol. Diagn. 20(4), 455–464 (2018).
    • 37. Arnold M , Sierra MS , Laversanne M , Soerjomataram I , Jemal A , Bray F . Global patterns and trends in colorectal cancer incidence and mortality. Gut 66(4), 683–691 (2017). • Describes the global trends in colorectal cancer incidence rates and mortality and highlights the differences between high- and low-income countries.