Targeted KRAS mutation assessment on patient tumor histologic material in real time diagnostics

Vassiliki Kotoula, Elpida Charalambous, Bart Biesmans, Andigoni Malousi, Eleni Vrettou, George Fountzilas, George Karkavelas, Vassiliki Kotoula, Elpida Charalambous, Bart Biesmans, Andigoni Malousi, Eleni Vrettou, George Fountzilas, George Karkavelas

Abstract

Background: Testing for tumor specific mutations on routine formalin-fixed paraffin-embedded (FFPE) tissues may predict response to treatment in Medical Oncology and has already entered diagnostics, with KRAS mutation assessment as a paradigm. The highly sensitive real time PCR (Q-PCR) methods developed for this purpose are usually standardized under optimal template conditions. In routine diagnostics, however, suboptimal templates pose the challenge. Herein, we addressed the applicability of sequencing and two Q-PCR methods on prospectively assessed diagnostic cases for KRAS mutations.

Methodology/principal findings: Tumor FFPE-DNA from 135 diagnostic and 75 low-quality control samples was obtained upon macrodissection, tested for fragmentation and assessed for KRAS mutations with dideoxy-sequencing and with two Q-PCR methods (Taqman-minor-groove-binder [TMGB] probes and DxS-KRAS-IVD). Samples with relatively well preserved DNA could be accurately analyzed with sequencing, while Q-PCR methods yielded informative results even in cases with very fragmented DNA (p<0.0001) with 100% sensitivity and specificity vs each other. However, Q-PCR efficiency (Ct values) also depended on DNA-fragmentation (p<0.0001). Q-PCR methods were sensitive to detect<or=1% mutant cells, provided that samples yielded cycle thresholds (Ct)<29, but this condition was met in only 38.5% of diagnostic samples. In comparison, FFPE samples (>99%) could accurately be analyzed at a sensitivity level of 10% (external validation of TMGB results). DNA quality and tumor cell content were the main reasons for discrepant sequencing/Q-PCR results (1.5%).

Conclusions/significance: Diagnostic targeted mutation assessment on FFPE-DNA is very efficient with Q-PCR methods in comparison to dideoxy-sequencing. However, DNA fragmentation/amplification capacity and tumor DNA content must be considered for the interpretation of Q-PCR results in order to provide accurate information for clinical decision making.

Conflict of interest statement

Competing Interests: The authors have declared that no competing interests exist.

Figures

Figure 1. FFPE DNA fragmentation and real…
Figure 1. FFPE DNA fragmentation and real time PCR method performance.
A: A typical series of diagnostic samples tested with the multiplex PCR assay for DNA fragmentation and KRAS exon 2 (intron spanning amplicon). Samples positive for products ≥300 bp are considered as good quality samples yielding the 340 bp product for exon 2 KRAS sequencing. In lanes 1, 3 and 4, faint KRAS product bands could be obtained from samples with very fragmented DNA but the corresponding capillary electropherograms were usually non-informative. In B, real time PCR efficiency largely depends on DNA fragmentation. DNA control Ct values reflect the amplification efficiency of FFPE DNA. With both TMGB and DxS-KRAS assays, Ct values from good quality samples were substantially lower than Ct values from very fragmented samples. The majority of FFPE DNA samples yielded DNA control Ct values between 29–33 (dotted lines). In C, typical results from diagnostic samples with DxS-KRAS. DNA control Cts for the standards contained in the kit are below 29 (arrow), but corresponding values for the diagnostic samples are >29.
Figure 2. Performance of a typical good…
Figure 2. Performance of a typical good quality FFPE-DNA diagnostic sample with the three methods applied for KRAS mutation assessment.
A and B: Corresponding tissue section with areas marked for macrodissection containing ∼70% tumor cells. Some necrotic areas can not be avoided but in this analogy these do not interfere with DNA extraction. The estimated number of sectioned neoplastic cells in B is ∼4000. For good quality samples, KRAS mutation assessment is reliable with any method, as shown in C (sequencing, c. 34 G>A corresponding to the G12S change), in D (TMGB-KRAS, G12S mutation positive) and in E (DxS-KRAS, G12S mutation positive). DNA control Ct values <29 (arrow) were yielded with both real time PCR methods (red curves in D and E).
Figure 3. KRAS-TMGB validation on good quality…
Figure 3. KRAS-TMGB validation on good quality FFPE DNA samples.
A. The method can provide reliable results with a broad range of DNA input, as shown here for a G12V mutant sample that was serially diluted up to 1.5 ng/reaction. dCts in all samples are kept close to 0 (safe mutation calling with this method). Red curves, DNA control; Green curves, G12V targeting assay. B. TMGB-KRAS was sensitive to detect <1% tumor cells in good quality FFPE-DNA samples. Seven different mutant FFPE samples containing ∼70% tumor cells (arrows) were diluted at 1∶10 and 1∶100 with different wild-type FFPE-DNA samples. Amplification curve Cts of the diluted mutant DNA showed some degree of linear increase in Ct values for the mutant target, as expected (diagonal lines).
Figure 4. Evaluation of KRAS-TMGB profiles.
Figure 4. Evaluation of KRAS-TMGB profiles.
As shown in A, mutant samples have significantly lower dCT values than non-mutant ones. The mutation status of KRAS codons 12 and 13 is evaluated based on the lowest value of the 7-dCt-profile for the corresponding mutant targets, provided that the lowest value falls below the cut-off in each case. In some cases, dCts for the non-mutant alleles can be very low, falling within the range of mutation calling, due to cross-reactivity (labeled as OTHER MUT). Such cross-reactions can be troublesome if evaluating each assay separately. By contrast, if evaluating the profile of dCt values for each sample, as shown in the two examples in B with TMGB, the mutant allele can readily be recognized. Here, on the left a sample with G12A, on the right a sample with G13D (duplicates). The dCt profiles of all samples analyzed in this study are shown in the colormap in C.
Figure 5. Typical example of a diagnostic…
Figure 5. Typical example of a diagnostic case with very low content in neoplastic cells.
If macrodissection is avoided in such cases, erroneous results are likely to be obtained. A, whole section of the CRC metastatic site (M) surrounded by normal liver (L). Circled areas are marked for macrodissection. In B, the metastatic site is largely composed of necrotic (N) and calcified (Ca++) elements within a loose stroma (s), while neoplastic cells (asterisks) correspond to <<1% in the whole section (A) and to ∼10% in the macrodissected areas (B). Two DNA samples were extracted from this specimen, one upon macrodissection and one from the whole section. As shown in C, although both DNA samples were of the same unfavorable quality (DNA control Ct∼34.5), it was possible to identify the G12D mutation in the macrodissected sample, while the sample obtained from the whole section appeared as wild type. Arrow in C, Ct = 29.

References

    1. Ciardiello F, Tortora G. EGFR antagonists in cancer treatment. N Engl J Med. 2008;358:1160–1174.
    1. Stewart DJ, Kurzrock R. Cancer: the road to Amiens. J Clin Oncol. 2009;27:328–333.
    1. Messersmith WA, Ahnen DJ. Targeting EGFR in colorectal cancer. N Engl J Med. 2008;359:1834–1836.
    1. Bach PB. Limits on Medicare's ability to control rising spending on cancer drugs. N Engl J Med. 2009;360:626–633.
    1. EMEA. EMEA/405113/2007. 2007.
    1. EMEA. EMEA/CHMP/280402/2008. 2008.
    1. De Roock W, Piessevaux H, De Schutter J, Janssens M, De Hertogh G, et al. KRAS wild-type state predicts survival and is associated to early radiological response in metastatic colorectal cancer treated with cetuximab. Ann Oncol. 2008;19:508–515.
    1. Karapetis CS, Khambata-Ford S, Jonker DJ, O'Callaghan CJ, Tu D, et al. K-ras mutations and benefit from cetuximab in advanced colorectal cancer. N Engl J Med. 2008;359:1757–1765.
    1. Lievre A, Bachet JB, Boige V, Cayre A, Le Corre D, et al. KRAS mutations as an independent prognostic factor in patients with advanced colorectal cancer treated with cetuximab. J Clin Oncol. 2008;26:374–379.
    1. Lievre A, Bachet JB, Le Corre D, Boige V, Landi B, et al. KRAS mutation status is predictive of response to cetuximab therapy in colorectal cancer. Cancer Res. 2006;66:3992–3995.
    1. Amado RG, Wolf M, Peeters M, Van Cutsem E, Siena S, et al. Wild-type KRAS is required for panitumumab efficacy in patients with metastatic colorectal cancer. J Clin Oncol. 2008;26:1626–1634.
    1. Frayling IM. Methods of molecular analysis: mutation detection in solid tumours. Mol Pathol. 2002;55:73–79.
    1. Hewitt SM, Lewis FA, Cao Y, Conrad RC, Cronin M, et al. Tissue handling and specimen preparation in surgical pathology: issues concerning the recovery of nucleic acids from formalin-fixed, paraffin-embedded tissue. Arch Pathol Lab Med. 2008;132:1929–1935.
    1. Paik S, Kim CY, Song YK, Kim WS. Technology insight: Application of molecular techniques to formalin-fixed paraffin-embedded tissues from breast cancer. Nat Clin Pract Oncol. 2005;2:246–254.
    1. von Ahlfen S, Missel A, Bendrat K, Schlumpberger M. Determinants of RNA quality from FFPE samples. PLoS ONE. 2007;2:e1261.
    1. Legrand B, Mazancourt P, Durigon M, Khalifat V, Crainic K. DNA genotyping of unbuffered formalin fixed paraffin embedded tissues. Forensic Sci Int. 2002;125:205–211.
    1. Andreyev HJ, Norman AR, Cunningham D, Oates JR, Clarke PA. Kirsten ras mutations in patients with colorectal cancer: the multicenter “RASCAL” study. J Natl Cancer Inst. 1998;90:675–684.
    1. Fearon ER, Vogelstein B. A genetic model for colorectal tumorigenesis. Cell. 1990;61:759–767.
    1. Pretlow TP, Brasitus TA, Fulton NC, Cheyer C, Kaplan EL. K-ras mutations in putative preneoplastic lesions in human colon. J Natl Cancer Inst. 1993;85:2004–2007.
    1. Jimeno A, Messersmith WA, Hirsch FR, Franklin WA, Eckhardt SG. KRAS Mutations and Sensitivity to Epidermal Growth Factor Receptor Inhibitors in Colorectal Cancer: Practical Application of Patient Selection. J Clin Oncol 2009
    1. van Krieken JH, Jung A, Kirchner T, Carneiro F, Seruca R, et al. KRAS mutation testing for predicting response to anti-EGFR therapy for colorectal carcinoma: proposal for an European quality assurance program. Virchows Arch. 2008;453:417–431.
    1. Cross J. DxS Ltd. Pharmacogenomics. 2008;9:463–467.
    1. Gallegos Ruiz MI, Floor K, Rijmen F, Grunberg K, Rodriguez JA, et al. EGFR and K-ras mutation analysis in non-small cell lung cancer: comparison of paraffin embedded versus frozen specimens. Cell Oncol. 2007;29:257–264.
    1. Lim EH, Zhang SL, Yu K, Nga ME, Ahmed DA, et al. An alternative approach to determining therapeutic choices in advanced non-small cell lung carcinoma (NSCLC): maximizing the diagnostic procedure and the use of low-volume lung biopsies. J Thorac Oncol. 2007;2:387–396.
    1. Morlan J, Baker J, Sinicropi D. Mutation detection by real-time PCR: a simple, robust and highly selective method. PLoS ONE. 2009;4:e4584.
    1. Rauser S, Weis R, Braselmann H, Feith M, Stein HJ, et al. Significance of HER2 low-level copy gain in Barrett's cancer: implications for fluorescence in situ hybridization testing in tissues. Clin Cancer Res. 2007;13:5115–5123.
    1. Brink M, de Goeij AF, Weijenberg MP, Roemen GM, Lentjes MH, et al. K-ras oncogene mutations in sporadic colorectal cancer in The Netherlands Cohort Study. Carcinogenesis. 2003;24:703–710.
    1. Andreyev HJ, Tilsed JV, Cunningham D, Sampson SA, Norman AR, et al. K-ras mutations in patients with early colorectal cancers. Gut. 1997;41:323–329.
    1. Burmer GC, Rabinovitch PS, Loeb LA. Frequency and spectrum of c-Ki-ras mutations in human sporadic colon carcinoma, carcinomas arising in ulcerative colitis, and pancreatic adenocarcinoma. Environ Health Perspect. 1991;93:27–31.
    1. van Dongen JJ, Langerak AW, Bruggemann M, Evans PA, Hummel M, et al. Design and standardization of PCR primers and protocols for detection of clonal immunoglobulin and T-cell receptor gene recombinations in suspect lymphoproliferations: report of the BIOMED-2 Concerted Action BMH4-CT98-3936. Leukemia. 2003;17:2257–2317.
    1. Johnson NA, Hamoudi RA, Ichimura K, Liu L, Pearson DM, et al. Application of array CGH on archival formalin-fixed paraffin-embedded tissues including small numbers of microdissected cells. Lab Invest. 2006;86:968–978.
    1. Kutyavin IV, Afonina IA, Mills A, Gorn VV, Lukhtanov EA, et al. 3′-minor groove binder-DNA probes increase sequence specificity at PCR extension temperatures. Nucleic Acids Res. 2000;28:655–661.
    1. Gibson NJ. The use of real-time PCR methods in DNA sequence variation analysis. Clin Chim Acta. 2006;363:32–47.
    1. Beeghly-Fadiel A, Long JR, Gao YT, Li C, Qu S, et al. Common MMP-7 polymorphisms and breast cancer susceptibility: a multistage study of association and functionality. Cancer Res. 2008;68:6453–6459.
    1. De la Vega FM, Lazaruk KD, Rhodes MD, Wenz MH. Assessment of two flexible and compatible SNP genotyping platforms: TaqMan SNP Genotyping Assays and the SNPlex Genotyping System. Mutat Res. 2005;573:111–135.
    1. Kim S, Misra A. SNP genotyping: technologies and biomedical applications. Annu Rev Biomed Eng. 2007;9:289–320.
    1. Morita A, Nakayama T, Doba N, Hinohara S, Mizutani T, et al. Genotyping of triallelic SNPs using TaqMan PCR. Mol Cell Probes. 2007;21:171–176.
    1. Letertre C, Perelle S, Dilasser F, Arar K, Fach P. Evaluation of the performance of LNA and MGB probes in 5′-nuclease PCR assays. Mol Cell Probes. 2003;17:307–311.
    1. Benlloch S, Paya A, Alenda C, Bessa X, Andreu M, et al. Detection of BRAF V600E mutation in colorectal cancer: comparison of automatic sequencing and real-time chemistry methodology. J Mol Diagn. 2006;8:540–543.
    1. Andreassen CN, Sorensen FB, Overgaard J, Alsner J. Optimisation and validation of methods to assess single nucleotide polymorphisms (SNPs) in archival histological material. Radiother Oncol. 2004;72:351–356.
    1. Cronin M, Pho M, Dutta D, Stephans JC, Shak S, et al. Measurement of gene expression in archival paraffin-embedded tissues: development and performance of a 92-gene reverse transcriptase-polymerase chain reaction assay. Am J Pathol. 2004;164:35–42.
    1. Itabashi T, Maesawa C, Uchiyama M, Higuchi T, Masuda T. Quantitative detection of mutant alleles of the K-ras gene with minor groove binder-conjugated fluorogenic DNA probes. Int J Oncol. 2004;24:687–696.
    1. Quach N, Goodman MF, Shibata D. In vitro mutation artifacts after formalin fixation and error prone translesion synthesis during PCR. BMC Clin Pathol. 2004;4:1.
    1. Marchetti A, Felicioni L, Buttitta F. Assessing EGFR mutations. N Engl J Med. 2006;354:526–528; author reply 526–528.
    1. Wang F, Wang L, Briggs C, Sicinska E, Gaston SM, et al. DNA degradation test predicts success in whole-genome amplification from diverse clinical samples. J Mol Diagn. 2007;9:441–451.
    1. Kosel S, Grasbon-Frodl EM, Arima K, Chimelli L, Hahn M, et al. Inter-laboratory comparison of DNA preservation in archival paraffin-embedded human brain tissue from participating centres on four continents. Neurogenetics. 2001;3:163–170.
    1. Ang PW, Toh HB, Iacopetta B, Soong R. An improved quality control for bisulfite-PCR-based DNA methylation analysis: cycle threshold value. Clin Chem Lab Med. 2008;46:1117–1121.
    1. Cukier HN, Pericak-Vance MA, Gilbert JR, Hedges DJ. Sample degradation leads to false-positive copy number variation calls in multiplex real-time polymerase chain reaction assays. Anal Biochem. 2009;386:288–290.
    1. Nielsen K, Mogensen HS, Hedman J, Niederstatter H, Parson W, et al. Comparison of five DNA quantification methods. Forensic Sci Int Genet. 2008;2:226–230.
    1. Do H, Krypuy M, Mitchell PL, Fox SB, Dobrovic A. High resolution melting analysis for rapid and sensitive EGFR and KRAS mutation detection in formalin fixed paraffin embedded biopsies. BMC Cancer. 2008;8:142.
    1. Tol J, Koopman M, Cats A, Rodenburg CJ, Creemers GJ, et al. Chemotherapy, bevacizumab, and cetuximab in metastatic colorectal cancer. N Engl J Med. 2009;360:563–572.
    1. Ogino S, Kawasaki T, Brahmandam M, Yan L, Cantor M, et al. Sensitive sequencing method for KRAS mutation detection by Pyrosequencing. J Mol Diagn. 2005;7:413–421.
    1. Ogino S, Nosho K, Kirkner GJ, Shima K, Irahara N, et al. PIK3CA mutation is associated with poor prognosis among patients with curatively resected colon cancer. J Clin Oncol. 2009;27:1477–1484.
    1. Packham D, Ward RL, Ap Lin V, Hawkins NJ, Hitchins MP. Implementation of novel pyrosequencing assays to screen for common mutations of BRAF and KRAS in a cohort of sporadic colorectal cancers. Diagn Mol Pathol. 2009;18:62–71.
    1. Di Nicolantonio F, Martini M, Molinari F, Sartore-Bianchi A, Arena S, et al. Wild-type BRAF is required for response to panitumumab or cetuximab in metastatic colorectal cancer. J Clin Oncol. 2008;26:5705–5712.
    1. Sartore-Bianchi A, Martini M, Molinari F, Veronese S, Nichelatti M, et al. PIK3CA mutations in colorectal cancer are associated with clinical resistance to EGFR-targeted monoclonal antibodies. Cancer Res. 2009;69:1851–1857.
    1. Zhu CQ, da Cunha Santos G, Ding K, Sakurada A, Cutz JC, et al. Role of KRAS and EGFR as biomarkers of response to erlotinib in National Cancer Institute of Canada Clinical Trials Group Study BR.21. J Clin Oncol. 2008;26:4268–4275.

Source: PubMed

Подписаться