Skip to main content
  • Technical advance
  • Open access
  • Published:

COLD-PCR enhanced melting curve analysis improves diagnostic accuracy for KRAS mutations in colorectal carcinoma

Abstract

Background

KRAS mutational analysis is the standard of care prior to initiation of treatments targeting the epidermal growth factor receptor (EGFR) in patients with metastatic colorectal cancer. Sensitive methods are required to reliably detect KRAS mutations in tumor samples due to admixture with non-mutated cells. Many laboratories have implemented sensitive tests for KRAS mutations, but the methods often require expensive instrumentation and reagents, parallel reactions, multiple steps, or opening PCR tubes.

Methods

We developed a highly sensitive, single-reaction, closed-tube strategy to detect all clinically significant mutations in KRAS codons 12 and 13 using the Roche LightCycler® instrument. The assay detects mutations via PCR-melting curve analysis with a Cy5.5-labeled sensor probe that straddles codons 12 and 13. Incorporating a fast COLD-PCR cycling program with a critical denaturation temperature (T c ) of 81°C increased the sensitivity of the assay >10-fold for the majority of KRAS mutations.

Results

We compared the COLD-PCR enhanced melting curve method to melting curve analysis without COLD-PCR and to traditional Sanger sequencing. In a cohort of 61 formalin-fixed paraffin-embedded colorectal cancer specimens, 29/61 were classified as mutant and 28/61 as wild type across all methods. Importantly, 4/61 (6%) were re-classified from wild type to mutant by the more sensitive COLD-PCR melting curve method. These 4 samples were confirmed to harbor clinically-significant KRAS mutations by COLD-PCR DNA sequencing. Five independent mixing studies using mutation-discordant pairs of cell lines and patient specimens demonstrated that the COLD-PCR enhanced melting curve assay could consistently detect down to 1% mutant DNA in a wild type background.

Conclusions

We have developed and validated an inexpensive, rapid, and highly sensitive clinical assay for KRAS mutations that is the first report of COLD-PCR combined with probe-based melting curve analysis. This assay significantly improved diagnostic accuracy compared to traditional PCR and direct sequencing.

Peer Review reports

Background

KRAS (K irsten ra t s arcoma virus homolog 2) is a membrane-anchored G-protein that acts downstream of the epidermal growth factor receptor (EGFR) to activate pro-growth and anti-apoptotic pathways, including the MAP kinase and PI3 kinase pathways [1]. Mutations in codons 12 and 13 of the KRAS gene confer resistance to drugs targeted at EGFR by impairing GTPase activity, which results in constitutive EGFR-independent signaling. KRAS is one of the most frequently mutated oncogenes in human cancer, with significant mutation rates in common epithelial malignancies such as colon cancer (~40%), lung cancer (~20%), gastric cancer (~10%), and pancreatic cancer (~65%) (COSMIC database; http://www.sanger.ac.uk/genetics/CGP/cosmic/) [2, 3].

The role of KRAS mutation status in clinical decision making is best defined for colon cancer. Several large randomized-controlled trials demonstrated no benefit from expensive anti-EGFR drugs such as cetuximab (Erbitux™) in patients with KRAS-mutant colon cancer [4, 5]. The National Comprehensive Cancer Network (NCCN) now recommends KRAS testing prior to initiation of anti-EGFR therapy in colon cancer patients [6]. Accumulating evidence also suggests a role for KRAS testing to guide therapy in patients with non-small cell lung cancer [7, 8].

Greater than 95% of KRAS mutations occur in codon 12 or codon 13. Within these codons G12 D (GG T to GA T), G12V (GG T to GT T), and G13 D (GG C to GA C) comprise ~80% of the mutations [2, 4]. Less frequent mutations include G12 S (G GT to A GT), G12C (G GT to T GT), G12R (G GT to C GT), and G12A (GG T to GC T). Silent mutations are exceedingly rare http://www.sanger.ac.uk/genetics/CGP/cosmic/.

CO-amplification at L ower D enaturation temperature (COLD)-PCR is a recently described method to selectively amplify mutant alleles in a wild type background that does not require any additional instrumentation or reagents to implement [9]. Two forms of COLD-PCR are described: fast COLD-PCR and full COLD-PCR (reviewed in [10]). Fast COLD-PCR enriches G:C to A:T mutations that slightly, but predictably lower the melting temperature (T m ) of the PCR amplicon, by using a critical denaturation temperature (T c ) that favors PCR amplification of the mutant allele. Full COLD-PCR theoretically enhances detection of any type of mutation via conditions which promote annealing of WT:mutant pairs and selective denaturation of these heteroduplexes at an empirically determined T c . As the name implies, fast COLD-PCR has the advantage of being more rapid than full COLD-PCR (1-2 hours of instrument time compared to 5-8 hours) and is also easier to troubleshoot and implement in our experience. Fast COLD-PCR is ideal for KRAS codon 12 and 13 mutational analysis because >90% of the mutations are G:C to A:T changes (G12 D, G12V, G12 S, G12C, G13D), and the remaining common mutations are T m -neutral G:C to G:C changes (G12R and G12A).

Here, we report a novel strategy for mutation detection that combines fast COLD-PCR with two-probe melting curve analysis using the LightCycler® instrument. By incorporating COLD-PCR we achieve >10-fold enhancement for the majority of KRAS mutations with a sensitivity of ~1% mutant allele in a wild type background.

Methods

Patient tumor samples and DNA extraction

All clinical specimens consisted of DNA prepared from formalin-fixed paraffin-embedded (FFPE) tissue from patients with sporadic colorectal carcinoma. De-identified residual clinical specimens were obtained in accordance with the declaration of Helsinki and ethics guidelines of the local institutional review board. We obtained a total of 61 de-identified specimens: 10 DNA specimens from the University of Texas MD Anderson Cancer Center, 4 specimens of unstained FFPE tissue sections with corresponding hematoxylin-eosin (HE) stained slides from the University of Chicago, and 47 DNA specimens prepared from FFPE tissue from colorectal cancer specimens at the University of Washington. HE-stained slides were used as a guide to manually dissect areas of tumor tissue from unstained slide sections. Genomic DNA was prepared with the Gentra Puregene DNA Isolation Kit (Qiagen, Cat. No. 158489).

KRAS mutational detection by Regular- and COLD-PCR enhanced melting curve analysis

Our basic melting-curve strategy to identify KRAS mutations is a modification of a procedure described by Nikiforova and colleagues [11]. The assay is a PCR fluorescence resonance energy transfer (FRET) hybridization probe assay performed with a LightCycler® carousel instrument (Roche Molecular Biochemicals, Manheim Germany). This format requires a pair of primers and two fluorescently labeled oligonucleotide probes and relies on FRET between two probes that hybridize adjacent to one another (Figure 1). For the detection of KRAS codon 12/13 mutations, the Cy5.5-labeled probe spans this region, and the products are distinguished based on their distinct T m , which reflects the thermodynamic stability of the perfectly complementary and mismatched probe-target duplexes. The wild-type T m is 69.5 ± 0.5°C, and the mutant T m is 62.8 ± 1°C. The assay cannot reliably determine the exact base change based on the T m . The primers and probes are given in Table 1. Each 20 μl PCR reaction contained 2 μl 10× LightCycler® FastStart DNAHyb Master mix (Roche Cat No. 03 002248 001), 0.5 units of LightCycler® uracil N-glycosylase enzyme (UNG) (Roche Cat No. 03539806001), 2 mM final MgCl2 concentration, 30 pmol KRAS-F (1.5 μM final concentration), 10 pmol KRAS-R (0.5 μM final concentration), 4 pmol anchor probe (0.2 μM final concentration), and 4 pmol sensor probe (0.2 μM final concentration). Template DNA was added at 10-200 ng per reaction. PCR reactions were set up in glass LightCycler® Capillaries (Roche Cat. No. 1 909 339) and run on both LightCycler® 1.5 and LightCycler® 2.0 carousel instruments. For the regular PCR melting curve assay (not COLD-PCR enhanced) the cycling program steps were as follows: 1) UNG step to remove carry-over amplicon, 40°C for 10 min, 2) activation of antibody-inhibited Taq polymerase, 94°C for 10 min, 3) cycling 94°C 1 s, 52°C 20 s, 72°C 10 s for 45 total cycles, 4) melting curve with continuous temperature ramping at 0.15°C/s from 45°C to 80°C, and 5) Cooling to 40°C. The COLD-PCR enhanced cycling program is shown in Table 2. An initial 10 cycles of regular PCR are performed to build up product, followed by 40 cycles of COLD-PCR with a critical denaturation temperature (T c ) of 81°C which favors amplification of mutant alleles. We determined the T c empirically by progressively lowering the denaturation temperature from 84°C → 82°C → 81°C → 80°C → 79°C. Melting curves were produced in both mutant and wild type samples down to T c = 80°C. We chose the T c at 81°C because the melting curves were difficult to interpret at T c = 80°C due to reduced PCR efficiency and increased signal-to-noise ratio (Additional file 1 Figure S1).

Figure 1
figure 1

Melting Curve Assay Design. PCR primers are designed within introns of the functional KRAS (Ki-Ras2) gene on chromosome 12 to avoid amplification of a KRAS (Ki-Ras1) pseudogene on chromosome 6. A Cy5.5-labeled wild type sensor probe sits over the mutation hotspot at codons 12 and 13. When a KRAS mutation is present, the sensor probe melts off at a lower T m .

Table 1 Primer and Probe Sequences (adapted from [11])
Table 2 Fast COLD-PCR Cycling Program

Regular- and COLD-PCR enhanced Sanger DNA sequencing

We used KRAS-F and KRAS-R primers (Table 1) for both PCR amplification and DNA sequencing. For DNA sequencing, regular- or COLD-PCR reactions were performed as described above except that the anchor and sensor probes were not added to the reactions and thermocycling was performed on a Bio-Rad C1000 instrument. PCR products were purified using DNA Clean & Concentrator™-5 (Zymo Research) and eluted in a 30 μl volume of elution buffer (Cat No D4003, D4004, D4013 & D4014). Each 10 μl sequencing reaction included 2 μl Big Dye Terminator v3.1 (Applied Biosystems, ABI) 2 μl 5× sequencing buffer (ABI), 4 pmol of sequencing primer (either KRAS-F or KRAS-R), and 200-300 ng purified PCR product. Sequencing reactions were cleaned up using AutoSeq G-50 Sephadex columns (Amersham Biosciences Cat. No. 27-5340), run on an ABI 3130 instrument, and analyzed with Mutation Surveyor software (Softgenetics).

We sequenced a total of 30 out of 61 samples including all 4 discrepant samples that were re-classified as KRAS-mutant by the COLD-PCR melting curve analysis (Additional file 2 Table S1). Two of the four discrepant samples had very low mutant peaks by COLD-PCR sequencing using the same thermocycling conditions (T c = 81°C, Table 2) that produced easily discernable mutant peaks in the COLD-PCR melting curve assay, presumably because the melting curve analysis is inherently more sensitive than sequencing. For this reason, we lowered the T c to 79.4°C to further enhance amplification of minority mutant alleles and confirm the KRAS mutations we detected in the 4 discrepant samples. We selected the T c of 79.4°C after we tried T c ranging from 79-81°C at 0.2°C increments and determined that the T c of 79.4°C was the lowest denaturation temperature in which PCR product from a wild type sample was still visible an agarose gel. We repeated COLD-PCR sequencing with T c = 79.4°C a total of 3 times for each of the 4 discrepant samples, obtaining concordant KRAS-mutant results and appropriately negative KRAS-wild type controls on each run.

Confirmation of Discrepant Specimens With An Alternative Assay

In addition to COLD-PCR sequencing, we confirmed the 4 discrepant specimens using the Lightmix KRAS mutation detection kit following the manufacturer's instructions (TIB Molbiol, Cat#04-0416-08). This assay uses a locked nucleic acid oligomer competitor and has a stated sensitivity of at least 1% for mutant allele in a wild type background (Additional file 1 Figure S2).

Mixing/Sensitivity Studies

We performed two types of mixing studies to estimate sensitivity: (1) KRAS-mutant colorectal cancer cell lines mixed with KRAS-WT peripheral blood leukocyte DNA, and (2) KRAS-mutant FFPE DNA mixed with KRAS-WT FFPE DNA. We used a total of three KRAS-mutant colorectal cancer cell lines, SW480 (homozygous G12V; GG T to GT T), HCT116 (heterozygous G13D; GG C to GA C) and LS174T (heterozygous G12D; GG T to GA T), and two KRAS-mutant FFPE colorectal cancer samples (one G12 D and one G13D). The undiluted G12 D and G13 D FFPE samples both had ~ 25% KRAS mutant allele based on a visual estimate of tumor nuclear content and on the mutant:WT peak height ratios in regular PCR melting curve analysis (data not shown). To control for potential differences in ploidy between samples and variation in DNA preparations that may confound the interpretation of mixing studies, we first adjusted each DNA preparation so that the cycle thresholds (Ct) of all mutant and WT samples were within 0.5 of each other (PCR-efficiency matched). Mutant cell lines or FFPE DNA samples were mixed in the following ratios (mutant%/WT%): 50/50, 20/80, 10/90, 5/95, 2/98, 1/99. We performed mixing studies in duplicate on a total of 5 unique mutant:WT pairs.

As an alternative measure of sensitivity, we tested 20% (G13C) and 1% (G12C) KRAS-mutant control DNA provided with the Lightmix KRAS mutation detection kit (TIB Molbiol, cat # 04-0416-08, Lot#4160902).

Results

Establishing and Validating COLD-PCR Conditions

To establish the critical denaturation temperature (T c ) for COLD-PCR melting curve analysis we selected a G12C KRAS-mutant FFPE colorectal cancer specimen with ~10% mutant allele based on a visual estimate of tumor nuclei and the mutant/WT melting peak height ratio using regular PCR. The mutant/WT peak height ratio increased by an average of 6.3-fold in this "low-positive" patient sample as the T c was lowered from 84°C → 82°C → 81°C (Figure 2). COLD-PCR DNA sequencing confirmed a proportional increase in the mutant T base at the first position of codon 12 (G12C = G GT→T GT) as the T c was lowered (Figure 2 insets). We found that using a T c lower than 81°C further enhanced the relative proportion of mutant allele, but reduced PCR efficiency to a degree that made it more difficult to interpret melting peaks (Additional file 1 Figure S1).

Figure 2
figure 2

Optimization of COLD-PCR Conditions. A G12C (G GT-T GT) KRAS-mutant colorectal cancer sample with approximately 10% mutant allele was tested at progressively lower critical denaturation temperatures (T c ). As the T c was progressively lowered, the height of the mutant melting peak at 62.5°C increased relative to the height of the wild-type melting peak at 70°C. DNA sequencing performed on COLD-PCR products corroborated an increase in the peak height of the mutant T base (c.34 G > T) at the first position of codon 12 (compare top and bottom panel insets).

We next validated the COLD-PCR melting curve assay using FFPE colorectal cancer specimens with distinct characterized KRAS mutations. COLD-PCR enhanced detection of G:C to A:T mutations (G12 D, G13 D, G12C, G12V) between 9.8 and 14.0 fold compared to regular PCR (Summarized in Table 3). Importantly, the COLD-PCR reaction conditions we established did not diminish detection of T m -neutral G:C to G:C mutations (G12R and G13R). Surprisingly, the fast COLD-PCR reaction conditions actually enhanced the detection of a G13R (G GC to C GC) mutation, which we did not expect to enhance based on a T m -neutral G > C change. We hypothesize that this enrichment is due to local sequence effects or to a small amount of WT:mutant heteroduplex formation during the primer annealing step, which favors amplification of mutant alleles by the same mechanism as full COLD-PCR (see Background for discussion of fast vs. full COLD-PCR) [10].

Table 3 COLD-PCR Enhancement According to Specific KRAS Mutation

Sensitivity of the COLD-PCR Enhanced Melting Curve Assay

To determine the sensitivity of the COLD-PCR melting curve assay to detect low quantities of mutant allele in WT background we adopted two complementary strategies: (1) we mixed KRAS-mutant cell line DNA with KRAS-WT peripheral blood leukocyte DNA, and (2) KRAS-mutant FFPE tumor DNA with KRAS-WT FFPE tumor DNA at mutant:WT ratios of 50/50, 20/80, 10/90, 5/95, 2/98, and 1/99. Prior to mixing we ensured the PCR-efficiency of each sample in a pair was matched within 0.5 Ct because variations in DNA quality, ploidy, and sample preparation may favor the amplification of one sample over the other and confound the estimation of sensitivity [12]. In a total of 5 unique sample pairings which included G12 D, G13 D, and G12V-mutant DNA, the assay could readily detect KRAS mutations down to 1% mutant allele in each case with COLD-PCR enhancement, but only down to 10% mutant allele with regular PCR (Figure 3).

Figure 3
figure 3

Sensitivity of the Assay. Five independent mixing studies were performed in duplicate using KRAS-mutant cell lines (SW480, LS174T, HCT116) paired with wild-type peripheral blood leukocyte DNA and KRAS-mutant formalin-fixed paraffin-embedded (FFPE) tissue samples paired with KRAS-WT FFPE samples as described in the methods section. For each mixing study the mutant melting peak height was measured as a percentage of the maximum mutant peak height observed in the undiluted KRAS-mutant sample specific to the particular Mutant:WT pairing. The left panel shows the average and SD (error bars) of the %mutant peak height across the 5 mixing studies using regular- (gray triangles) or COLD-PCR (black squares) conditions. Mutant peak heights were consistently above the limit of detection down to 1% mutant allele using COLD-PCR compared to 10% mutant allele with regular PCR. The right panel shows an example of one of the mixing studies (SW480 cell line; homozygous G12V) used to generate the sensitivity graph in the left panel. Mutant melting peaks are not clearly discernible for 1%, 2%, and 5% KRAS-mutant samples using regular PCR (top right panel), but are readily detected using COLD-PCR (bottom right panel).

As a second measure of sensitivity, we ran commercial 20% (G13C) and 1% (G12C) KRAS-mutant control DNA (TIB Molbiol, Lot No. 4160902). The COLD-PCR enhanced melting curve assay correctly classified both controls as KRAS-mutant with a mutant:WT peak height ratio of 3.5 for the 20% G13C control, and 0.3 for the 1% G12C control (Additional file 1 Figure S3).

We next assessed if the COLD-PCR enhanced melting curve assay was sensitive enough to eliminate the need for tumor dissection in a KRAS-mutant sample with a low percentage (~3%) of tumor cells. We confirmed that both regular-PCR melting curve analysis and Sanger sequencing detected a KRAS mutation (G12D) when DNA was prepared with morphology-guided dissection to enrich for tumor cells in our standard fashion. We then tested a DNA sample prepared from the same tumor specimen without dissection (scraping the whole slide). The COLD-PCR enhanced melting curve assay readily detected the KRAS mutation when the whole slide was scraped, whereas the result with regular PCR melting curve analysis was equivocal (Figure 4).

Figure 4
figure 4

KRAS Mutation Detection Without Dissection. DNA was prepared from a G12 D KRAS-mutant colorectal cancer resection specimen with ~3% tumor cells by scraping the tissue from the entire slide without tumor dissection. The KRAS result was equivocal using regular-PCR (upper left panel), but a mutation was clearly detected using COLD-PCR (lower left panel). The corresponding HE-stained slide and region of tumor are depicted in the right panel.

COLD-PCR Enhanced Melting Curve Analysis Detects Mutations That Are Missed By Regular PCR and Direct Sanger Sequencing

We compared the COLD-PCR melting curve assay to regular PCR melting analysis in a cohort of 61 FFPE colorectal cancer specimens which included 14 blinded samples that had KRAS testing done previously at outside institutions. KRAS mutations were detected in 29/61(48%) samples by both regular- and COLD PCR melting curve analysis and, importantly, in an additional 4 samples (33/61 total; 54%) by COLD-PCR enhanced melting curve analysis only (Table 4; Additional file 2 Table S1). Regular Sanger sequencing did not detect mutations in the 4 samples that were re-classified from KRAS-WT to KRAS-mutant by COLD-PCR, presumably because the mutant allele % was below the limit of detection (sensitivity ~20 percent for standard sequencing). However, COLD-PCR enhanced DNA sequencing confirmed that the 4 re-classified samples each harbored a clinically significant KRAS mutation (Additional file 3; Figure 5; see methods section for a detailed discussion of COLD-PCR sequencing). KRAS mutations in the 4 re-classified samples were also confirmed using the TIB Molbiol Lightmix assay, which has a stated sensitivity of at least 1% (Additional file 1 Figure S2). Sequencing of an additional 26 specimens that were concordant by regular- and COLD-PCR confirmed the KRAS melting curve analysis result in all cases (Additional file 2 Table S1).

Table 4 Comparison of Regular- and COLD-PCR in 61 FFPE Samples
Figure 5
figure 5

Re-classified specimen. Shown are results from one of the 4 specimens that was re-classified from KRAS-WT to KRAS-mutant by COLD-PCR (sample #3 in Additional file 3). With regular PCR no definite mutant allele is detected by the melting curve assay, or by direct sequencing (left panel). Using COLD-PCR a clear mutant peak is visible in the melting curve assay and confirmed as a G12C mutation by sequencing (c.34 G > C). Arrows point to the position of the mutant melting peak and mutant base pair position. Note that a T c of 79.4°C was used for COLD-PCR sequencing compared to a T c of 81°C for the COLD-PCR melting curve assay (see Methods), which is the likely explanation for the higher proportion of mutant allele observed by COLD-PCR sequencing (bottom right).

Among the 14 specimens from outside institutions, KRAS results were concordant using the less sensitive regular PCR melting curve analysis and Sanger sequencing (7/14 KRAS-mutant; 7/14 KRAS-WT). However, we re-classified 1/14 of these specimens as KRAS-mutant by both COLD-PCR melting curve analysis and COLD-PCR sequencing. The method used by the outside laboratory to identify KRAS mutations was regular sequencing, which explains why the KRAS mutation in that specimen was not originally detected.

Discussion

Cancer tissue presents challenges for KRAS mutation analysis because each specimen is a unique mixture of tumor and non-tumor cells. A typical formalin-fixed paraffin-embedded colorectal cancer specimen might have between 2-50% tumor cells, which corresponds to 1-25% mutant allele when the tumor is diploid and a heterozygous mutation is present. To enrich the proportion of tumor cells most laboratories (including ours) manually dissect areas of tumor tissue from unstained sections guided by an HE-stained slide. This approach improves mutation detection, but tumors with a diffuse growth pattern or dense inflammatory infiltrate may still have a significant proportion of "contaminating" non-tumor DNA after tumor dissection. For this reason, methods such as COLD-PCR and peptide nucleic acid (PNA) clamping have been developed to increase the sensitivity of mutation analysis by selectively amplifying mutant alleles in a wild type background [9, 10, 13].

Here, we demonstrate that COLD-PCR combined with probe-based melting curve analysis can reproducibly detect down to 1% KRAS-mutant allele in a wild type background. This is comparable to the sensitivity reported with PNA clamping methods applied to melting curve analysis [14, 15]. We show that our COLD-PCR method can readily detect the 20% and 1% KRAS-mutant controls used in the nucleic acid clamping-based TIB Molbiol commercial kit for the LightCycler® instrument, with substantially less expensive reagents (~$5 compared to ~$100 per reaction). Limitations of our study include the relatively small sample size (n = 61), and lack of validation of the full range of rare KRAS mutations. A larger scale study is required to determine the assay's performance on the full complement of rare KRAS mutations.

A recent editorial on COLD-PCR technology suggested that "The utility of COLD-PCR will be greatly enhanced if this approach can be demonstrated to completely eliminate microdissection steps or at least to substantially reduce the tumor percentage required....[16]." We show that the COLD-PCR melting curve assay is sensitive enough to readily detect a KRAS mutation without tumor dissection in a sample with only ~3% tumor cells. Despite this, we believe it is prudent to continue dissection for tumor enrichment because additional mutational testing may be required on the same DNA preparation, and these add-on tests may have lower sensitivity.

Among a cohort of 61 sporadic colorectal cancer specimens we found that 4/33 (12.1%) specimens harboring KRAS mutations were incorrectly classified as KRAS-WT using regular (conventional) PCR melting curve analysis and Sanger sequencing, despite dissection of samples for tumor enrichment. An additional 10/33 KRAS-mutant specimens were close to the limit of detection using standard melting curve analysis (Table 4 "Lo-Pos"). These findings are supported by recent reports of false negative or equivocal KRAS results when less sensitive methods are used [13, 17, 18]. One study identified 10/57 (17.5%) additional KRAS-mutant samples when COLD-PCR was incorporated into high resolution melting curve analysis [17]. Kobunai and colleagues detected KRAS mutations in an additional 25 out of 224 colorectal tumors when a highly sensitive PNA-clamping method was compared to direct sequencing [13]. A third group reported equivocal KRAS results in 10% (18/180) of FFPE tumor specimens using a melting curve approach that was similar to our assay without COLD-PCR enhancement [18].

Misclassification of KRAS mutation status has important clinical consequences because KRAS testing of tumor tissue is often used as the sole determinant for selection of anti-EGFR therapy [19, 20]. Taken together, our data and the results of other recent studies suggest that more than 10% of patients with KRAS codon 12/13 mutations are incorrectly classified as wild type when less sensitive methods are used. This has implications for patient care and for the interpretation of results from clinical trials that assessed KRAS mutations by direct sequencing, such as the 2008 landmark New England Journal of Medicine study by Karapetis and colleagues [4].

Previous studies have reported COLD-PCR applied to KRAS mutational analysis [9, 17, 21] and COLD-PCR combined with high-resolution melting curve analysis [17, 22], but to our knowledge this is the first report of COLD-PCR combined with fluorescent probe-based melting analysis. Mancini and colleagues applied COLD-PCR to KRAS mutation detection using the Corbett Research RotorGene 6000 high resolution melting instrument [17]. The method described by Zuo et. al. 2009 utilized a ~7 hour full COLD-PCR protocol followed by a second reaction in a pyrosequencing instrument and achieved 1.1 to 4.7-fold enhancement KRAS-mutant signal compared to regular PCR [21]. Milbury and colleagues used single-step and nested PCR conditions followed by high resolution melting curve in a separate instrument [22]. There are important advantages to the novel strategy we describe here which make it appealing for the clinical setting compared to earlier reports. Most importantly, our method is performed as a single ~1 hour reaction and is entirely closed-tube, which cuts down substantially on labor and instrument time, while minimizing the risk of sample handling errors and contamination. Additionally, compared to high resolution melting curve analysis, our probe-based method offers theoretical increased specificity afforded by 2 primers and 2 probes (compared to 2 primers only), and better suitability for laboratories that already own or have experience with the Roche LightCycler® instrument.

Conclusion

We report a rapid, single-reaction, closed-tube clinical assay for KRAS mutations that combines COLD-PCR with melting curve analysis using the LightCycler® instrument. The assay has a sensitivity of at least 1%, which is comparable to or better than many commercial kits and previous studies, although methods with superior sensitivity have been reported [2224]. In a cohort of 61 colorectal cancer samples, the assay improved diagnostic accuracy by correctly identifying 4 KRAS-mutant samples that were not detected by regular melting curve analysis or direct sequencing. Incorporation of COLD-PCR with probe-based melting curve analysis was straight-forward to optimize and implement, and is therefore an appealing strategy for assays designed to detect low-level minority alleles.

Abbreviations

KRAS :

K irsten ra t s arcoma virus homolog 2

EGFR :

Epidermal growth factor receptor

COLD-PCR :

Co-amplification at lower denaturation temperature polymerase chain reaction

FFPE :

Formalin-fixed paraffin-embedded

WT :

Wild type

T m :

Melting temperature

T c :

Critical denaturation temperature

Ct :

Cycle Threshold

PNA :

Peptide nucleic acid

References

  1. Siena S, Sartore-Bianchi A, Di Nicolantonio F, Balfour J, Bardelli A: Biomarkers predicting clinical outcome of epidermal growth factor receptor-targeted therapy in metastatic colorectal cancer. J Natl Cancer Inst. 2009, 101 (19): 1308-1324. 10.1093/jnci/djp280.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Bamford S, Dawson E, Forbes S, Clements J, Pettett R, Dogan A, Flanagan A, Teague J, Futreal PA, Stratton MR, et al: The COSMIC (Catalogue of Somatic Mutations in Cancer) database and website. Br J Cancer. 2004, 91 (2): 355-358.

    CAS  PubMed  PubMed Central  Google Scholar 

  3. Forbes SA, Tang G, Bindal N, Bamford S, Dawson E, Cole C, Kok CY, Jia M, Ewing R, Menzies A: COSMIC (the Catalogue of Somatic Mutations in Cancer): a resource to investigate acquired mutations in human cancer. Nucleic Acids Res. 2010, D652-657. 10.1093/nar/gkp995. 38 Database

  4. Karapetis CS, Khambata-Ford S, Jonker DJ, O'Callaghan CJ, Tu D, Tebbutt NC, Simes RJ, Chalchal H, Shapiro JD, Robitaille S, et al: K-ras mutations and benefit from cetuximab in advanced colorectal cancer. N Engl J Med. 2008, 359 (17): 1757-1765. 10.1056/NEJMoa0804385.

    Article  CAS  PubMed  Google Scholar 

  5. Van Cutsem E, Kohne CH, Hitre E, Zaluski J, Chang Chien CR, Makhson A, D'Haens G, Pinter T, Lim R, Bodoky G, et al: Cetuximab and chemotherapy as initial treatment for metastatic colorectal cancer. N Engl J Med. 2009, 360 (14): 1408-1417. 10.1056/NEJMoa0805019.

    Article  CAS  PubMed  Google Scholar 

  6. Pritchard CC, Grady WM: Colorectal cancer molecular biology moves into clinical practice. Gut. 2010,

    Google Scholar 

  7. Linardou H, Dahabreh IJ, Bafaloukos D, Kosmidis P, Murray S: Somatic EGFR mutations and efficacy of tyrosine kinase inhibitors in NSCLC. Nat Rev Clin Oncol. 2009, 6 (6): 352-366. 10.1038/nrclinonc.2009.62.

    Article  CAS  PubMed  Google Scholar 

  8. Massarelli E, Varella-Garcia M, Tang X, Xavier AC, Ozburn NC, Liu DD, Bekele BN, Herbst RS, Wistuba II: KRAS mutation is an important predictor of resistance to therapy with epidermal growth factor receptor tyrosine kinase inhibitors in non-small-cell lung cancer. Clin Cancer Res. 2007, 13 (10): 2890-2896. 10.1158/1078-0432.CCR-06-3043.

    Article  CAS  PubMed  Google Scholar 

  9. Li J, Wang L, Mamon H, Kulke MH, Berbeco R, Makrigiorgos GM: Replacing PCR with COLD-PCR enriches variant DNA sequences and redefines the sensitivity of genetic testing. Nat Med. 2008, 14 (5): 579-584. 10.1038/nm1708.

    Article  CAS  PubMed  Google Scholar 

  10. Li J, Makrigiorgos GM: COLD-PCR: a new platform for highly improved mutation detection in cancer and genetic testing. Biochem Soc Trans. 2009, 37 (Pt 2): 427-432. 10.1042/BST0370427.

    Article  CAS  PubMed  Google Scholar 

  11. Nikiforova MN, Lynch RA, Biddinger PW, Alexander EK, Dorn GW, Tallini G, Kroll TG, Nikiforov YE: RAS point mutations and PAX8-PPAR gamma rearrangement in thyroid tumors: evidence for distinct molecular pathways in thyroid follicular carcinoma. J Clin Endocrinol Metab. 2003, 88 (5): 2318-2326. 10.1210/jc.2002-021907.

    Article  CAS  PubMed  Google Scholar 

  12. Ogino S, Kawasaki T, Brahmandam M, Yan L, Cantor M, Namgyal C, Mino-Kenudson M, Lauwers GY, Loda M, Fuchs CS: Sensitive sequencing method for KRAS mutation detection by Pyrosequencing. J Mol Diagn. 2005, 7 (3): 413-421.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Kobunai T, Watanabe T, Yamamoto Y, Eshima K: The frequency of KRAS mutation detection in human colon carcinoma is influenced by the sensitivity of assay methodology: a comparison between direct sequencing and real-time PCR. Biochem Biophys Res Commun. 2010, 395 (1): 158-162. 10.1016/j.bbrc.2010.03.167.

    Article  CAS  PubMed  Google Scholar 

  14. Oh JE, Lim HS, An CH, Jeong EG, Han JY, Lee SH, Yoo NJ: Detection of low-level KRAS mutations using PNA-mediated asymmetric PCR clamping and melting curve analysis with unlabeled probes. J Mol Diagn. 2010, 12 (4): 418-424. 10.2353/jmoldx.2010.090146.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Whitehall V, Tran K, Umapathy A, Grieu F, Hewitt C, Evans TJ, Ismail T, Li WQ, Collins P, Ravetto P, et al: A multicenter blinded study to evaluate KRAS mutation testing methodologies in the clinical setting. J Mol Diagn. 2009, 11 (6): 543-552. 10.2353/jmoldx.2009.090057.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Luthra R, Zuo Z: COLD-PCR finds hot application in mutation analysis. Clin Chem. 2009, 55 (12): 2077-2078. 10.1373/clinchem.2009.136143.

    Article  CAS  PubMed  Google Scholar 

  17. Mancini I, Santucci C, Sestini R, Simi L, Pratesi N, Cianchi F, Valanzano R, Pinzani P, Orlando C: The Use of COLD-PCR and High-Resolution Melting Analysis Improves the Limit of Detection of KRAS and BRAF Mutations in Colorectal Cancer. J Mol Diagn. 2010, 12 (5): 705-711. 10.2353/jmoldx.2010.100018.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Tsiatis AC, Norris-Kirby A, Rich RG, Hafez MJ, Gocke CD, Eshleman JR, Murphy KM: Comparison of Sanger sequencing, pyrosequencing, and melting curve analysis for the detection of KRAS mutations: diagnostic and clinical implications. J Mol Diagn. 2010, 12 (4): 425-432. 10.2353/jmoldx.2010.090188.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. European Medicines Agency: Committee for Medicinal Products for Human Use May 2008 Plenary Meeting monthly report. [http://www.emea.europa.eu/pdfs/human/press/pr/27923508en.pdf]

  20. National Comprehensive Cancer Network (NCCN) Clinical Practice Guidelines in Oncology: Colon Cancer V.2.2010. [http://www.nccn.org/professionals/physician_gls/PDF/colon.pdf]

  21. Zuo Z, Chen SS, Chandra PK, Galbincea JM, Soape M, Doan S, Barkoh BA, Koeppen H, Medeiros LJ, Luthra R: Application of COLD-PCR for improved detection of KRAS mutations in clinical samples. Mod Pathol. 2009, 22 (8): 1023-1031. 10.1038/modpathol.2009.59.

    Article  CAS  PubMed  Google Scholar 

  22. Milbury CA, Li J, Makrigiorgos GM: COLD-PCR-enhanced high-resolution melting enables rapid and selective identification of low-level unknown mutations. Clin Chem. 2009, 55 (12): 2130-2143. 10.1373/clinchem.2009.131029.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Kristensen LS, Daugaard IL, Christensen M, Hamilton-Dutoit S, Hager H, Hansen LL: Increased sensitivity of KRAS mutation detection by High-Resolution Melting analysis of COLD-PCR products. Hum Mutat. 2010, epub

    Google Scholar 

  24. Luo JD, Chan EC, Shih CL, Chen TL, Liang Y, Hwang TL, Chiou CC: Detection of rare mutant K-ras DNA in a single-tube reaction using peptide nucleic acid as both PCR clamp and sensor probe. Nucleic Acids Res. 2006, 34 (2): e12-10.1093/nar/gnj008.

    Article  PubMed  PubMed Central  Google Scholar 

Pre-publication history

Download references

Acknowledgements

We thank UT MD Anderson Cancer Center Molecular Diagnostic Laboratory for providing de-identified clinical specimens. We thank Dr. Deborah Barden and Dr. Karen Stephens for help in designing experiments and preparing the manuscript. We thank Dr. William Grady for providing colorectal cancer cell line DNA

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Colin C Pritchard.

Additional information

Competing interests

The authors declare that they have no competing interests.

Authors' contributions

CP developed the assay, designed the study, helped perform the experiments, analyzed the data, and drafted the manuscript. LA helped perform the experiments. PR and LJ helped design the study, provided FFPE specimens, commercial controls, scientific guidance, and reviewed the manuscript. JT helped design the study, analyze the data, review the manuscript, and also provided resources and scientific guidance. All authors have read and approved the final manuscript.

Electronic supplementary material

Additional file 1:Supplemental Figures S1-S3. Supplemental figures S1, S2, and S3, with figure legends. (PDF 403 KB)

12907_2010_78_MOESM2_ESM.PDF

Additional file 2:Table S1. Supplemental Table 1: Complete list of mutations detected by regular- or COLD-PCR. (PDF 42 KB)

12907_2010_78_MOESM3_ESM.PDF

Additional file 3:Sequencing results on the re-classified specimens. Regular- and COLD-PCR sequencing data for the 4 re-classified (discrepant) specimens. (PDF 157 KB)

Authors’ original submitted files for images

Rights and permissions

This article is published under license to BioMed Central Ltd. This is an Open Access article distributed under the terms of the Creative Commons Attribution License (http://creativecommons.org/licenses/by/2.0), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.

Reprints and permissions

About this article

Cite this article

Pritchard, C.C., Akagi, L., Reddy, P.L. et al. COLD-PCR enhanced melting curve analysis improves diagnostic accuracy for KRAS mutations in colorectal carcinoma. BMC Clin Pathol 10, 6 (2010). https://doi.org/10.1186/1472-6890-10-6

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/1472-6890-10-6

Keywords