Skip to main content
Biology Methods & Protocols logoLink to Biology Methods & Protocols
. 2024 Jun 4;9(1):bpae041. doi: 10.1093/biomethods/bpae041

PROMER technology: A new real-time PCR tool enabling multiplex detection of point mutation with high specificity and sensitivity

Hwanhee Nam 1, Esder Lee 2, Hichang Yang 3, Kyeyoon Lee 4, Taeho Kwak 5, Dain Kim 6, Hyemin Kim 7, Mihwa Yang 8, Younjoo Yang 9, Seungwan Son 10, Young-Hyean Nam 11, Il Minn 12,13,
PMCID: PMC11208725  PMID: 38938409

Abstract

Real-time polymerase chain reaction (real-time PCR) is a powerful tool for the precise quantification of nucleic acids in various applications. In cancer management, the monitoring of circulating tumor DNA (ctDNA) from liquid biopsies can provide valuable information for precision care, including treatment selection and monitoring, prognosis, and early detection. However, the rare and heterogeneous nature of ctDNA has made its precise detection and quantification challenging, particularly for ctDNA containing hotspot mutations. We have developed a new real-time PCR tool, PROMER technology, which enables the precise and sensitive detection of ctDNA containing cancer-driven single-point mutations. The PROMER functions as both a PRObe and priMER, providing enhanced detection specificity. We validated PROMER technology using synthetic templates with known KRAS point mutations and demonstrated its sensitivity and linearity of quantification. Using genomic DNA from human cancer cells with mutant and wild-type KRAS, we confirmed that PROMER PCR can detect mutant DNA. Furthermore, we demonstrated the ability of PROMER technology to efficiently detect mutation-carrying ctDNA from the plasma of mice with human cancers. Our results suggest that PROMER technology represents a promising new tool for the precise detection and quantification of DNA containing point mutations in the presence of a large excess of wild-type counterpart.

Keywords: real-time PCR, circulating tumor DNA, cell-free DNA, KRAS mutations, liquid biopsy

Introduction

Cell-free DNA (cfDNA) is a mixture of DNA fragments found in bodily fluids, originating from host cells in the process of their turnover [1, 2]. cfDNA can contain disease-specific DNA from the host, such as circulating tumor DNA (ctDNA) in the case of cancer patients [3]. Real-time monitoring and analysis of ctDNA has yielded valuable insights for personalized cancer care, including early detection [4–7], assessment of treatment efficacy [8, 9], detection of minimal residual disease [10–13], and identification of drug targets [14, 15] or resistance [16, 17]. ctDNA contains informative features such as genetic variation, copy number variation, fragmentation and rearrangement, and unique epigenetic aberrations. Among these characteristics, cancer-specific point mutations are the first and most used for approved companion diagnostics, such as Epidermal Growth Factor Receptor for non-small cell lung cancer and PIK3CA for breast cancer [5, 11, 18, 19]. Other targets, including ALK, ATM, BRACA1/2, BRAF, KRAS, MET, RET, and ROS1 are undergoing clinical evaluation [20]. Some studies examined a number of known mutations to establish a threshold for tumor detectability from single patient [11, 21, 22]. Together, ctDNA presents promising features for precision health applications.

Despite its potential, the precise and quantitative detection of ctDNA from cfDNA in plasma remains a challenging task. While the concentration of cfDNA in the plasma of cancer patients is higher than that of healthy individuals, the proportion of ctDNA [variant allele frequency (VAF)] in the isolated cfDNA varies significantly for different types of cancer and among patients with the same cancer [23, 24]. ctDNA is believed to be released through the apoptotic or necrotic turnover of cancer cells. However, the rate of cell death varies among different types of cancer, and the background levels of cfDNA originating from hematopoietic cells also vary among individuals [25, 26]. Intra-patient tumor heterogeneity can also complicate the detection of mutant-harboring ctDNA, as its quantity can be extremely low or it may exist in multiple forms of mutation [27, 28]. Furthermore, cfDNA has a rapid turnover, with a reported half-life ranging from 16 to 150 min [23, 29]. These characteristics necessitate the development of fast, highly sensitive, and precise detection tools for the analysis of ctDNA, particularly for the identification of cancer-specific point mutations.

Sequencing, quantitative polymerase chain reaction (PCR), and mass spectrometry have been the techniques widely used for the detection of ctDNA from patients’ liquid biopsies. Each technique offers specialized and in-depth analysis based on its unique principle. Whole-genome or whole-exome sequencing via next-generation sequencing (NGS) can provide comprehensive information on genetic variations [30, 31]. However, the low sensitivity of this approach makes it unsuitable for detecting rare ctDNA with <0.1% VAF. Additionally, NGS is not cost- or time-effective for clinical scenarios requiring rapid analysis. Several targeted sequencing approaches have been developed to investigate only certain loci of interest. These include tagged-amplicon deep sequencing (Tam-Seq) [32], Safe-Sequencing System (Safe-SeqS) [33], simple, multiplexed, PCR-based barcoding of DNA for sensitive mutation detection using sequencing (SiMSen-seq) [34], and cancer personalized profiling by deep sequencing (CAPP-Seq) [21]. PCR-based assays generally achieve enhanced sensitivity and precise quantification. Digital PCR technology such as droplet digital PCR (ddPCR) and BEAMing (beads emulsion amplification and magnetics) have been developed for ctDNA detection [35]. Real-time PCR provides a cost-effective, sensitive, and fast tool for ctDNA detection, especially for mutations with low allele frequency. Advanced real-time PCR such as allele-specific amplification (AS-PCR) [36] and co-amplification at lower denaturation temperature (COLD-PCR) [37] have been developed. Despite these technical advancements in improving detection efficacy and lowering error rates, continued research in ctDNA detection technology for diversified and improved performance is warranted for faster clinical implementation of ctDNA.

In this article, we present a new real-time PCR tool, named as PROMER technology [38], which has been developed for the sensitive and precise detection of small variations in DNA, such as single-point mutations, within a mixture of nucleic acids. We have validated the efficacy of PROMER technology using both synthetic DNA templates and cancer genomic DNA. Additionally, we have tested the technology in a multiplexed assay format. Finally, we have evaluated its capability to detect ctDNA in the blood of cancer-bearing animal models.

Material and methods

Reagents for PCR

The KRAS synthetic templates (Supplementary Table S1), PROMERs, and primers (Supplementary Table S2) were custom synthesized by Integrated DNA Technologies (Newark, NJ). KRAS TaqMan® Mutation Detection Assays (Assay IDs: Hs00000113_mu, Hs00000113_mu, Hs00000117_mu, Hs00000119_mu, Hs00000121_mu, Hs00000123_mu, Hs00000125_mu, Hs00000131_mu, Hs00000137_mu, and Hs00000139_mu) were purchased from Applied Biosystems (Waltham, MA, USA). RNase H2 was purchased from BioAssay Co. Ltd (Daejeon, South Korea) or custom produced. Briefly, cDNA of RNase H2 (pyrococcus furiosus, GenBank: CP023154.1) was cloned into pET-28a plasmid and transfected into BL21(DE3)pLysS strain. The expression of the RNase H2 was induced with IPTG and purified using Ni-NTA affinity chromatography (ThermoFisher Scientific, Waltham, MA, USA). 5x Apta Taq DNA Master was purchased from Roche (Basel, Switzerland).

Cell lines and genomic DNA preparation

The cell lines listed in Supplementary Table S3 were purchased from the American Type Culture Collection (ATCC, Manassas, Virginia) and were maintained according to the provider’s instructions. Genomic DNA from the cell lines was extracted using the Quick-DNA Miniprep Plus kit (Zymo Research, Irvine, CA, USA) according to the manufacturer’s instructions. Isolated genomic DNA was quantified using a NanoDrop™ Lite (ThermoFisher Scientific, Waltham, MA, USA).

Animals study

The animal experiments were conducted in accordance with a protocol approved by the Johns Hopkins Animal Care and Use Committee, and in compliance with the regulations of the Animal Welfare Act and Public Health Service (PHS) Policy. Johns Hopkins University has an approved PHS assurance. NSG (NOD/SCID/IL2Rγnull) mice were obtained from the Animal Resources Core of the Sydney Kimmel Comprehensive Cancer Center at Johns Hopkins University. NSG mice were administered with 1 million cells on their lower left flank. Tumor sizes were recorded twice a week. The animals were observed daily for changes in weight and any abnormalities. Animals were euthanized using a CO2 chamber, and whole blood was collected in 1.7 ml tube supplemented with 180 µl of acid citrate dextrose (ACD) solution (Becton Dickinson, Franklin Lakes, NJ, USA) for the isolation of cfDNA. The cfDNA was isolated using the MagMax Cell-Free DNA Isolation kit (Applied Biosystems, Waltham, MA, USA). The length and quantity of the isolated cfDNA were measured using the Agilent 2100 BioAnalyzer (Agilent, Santa Clara, CA, USA) at the Johns Hopkins Sidney Kimmel Comprehensive Cancer Center Experimental and Computational Genomics Core.

PCR reactions

PCR reactions were performed using QuantStudio 3, QuantStudio 5, QuantStuido 12K Flex (Applied Biosystems, Waltham, MA, USA), or CFX96 Touch Real-Time PCR Detection System (Bio-Rad, Hercules, CA, USA). Detailed PCR setup and thermal-cycling conditions are listed in Supplementary Table S4.

Results

PROMER technology for precise and sensitive detection of single-point mutation

We developed a new real-time PCR tool named PROMER (a combination of PRObe and priMER) technology designed for the precise and sensitive detection of single-point mutation. The PROMER is composed of a DNA–RNA–DNA hybrid oligonucleotide that has a fluorophore attached to its 5′-end and a quencher attached to its 3′-end (Fig. 1A). The DNA bases at the 5′-end of the PROMER serve as an extension group that acts as a 5′-primer for the PCR reaction. The cleavage group, which consists of 1 or 2 RNA bases, provides detection specificity by enabling a type 2 Ribonuclease H (RNase H2) to cut the phosphodiester bond of RNA in the DNA: RNA double strand, leaving a 3′-hydroxyl group. This cleavage only takes place when the DNA: RNA base pairing is an exact match [39, 40]. The principle of integrating RNase H2 into PCR (rhPCR) has been previously explored and demonstrated to enhance the specificity of PCR detection by mitigating off-target amplifications [41]. This study utilized two sets of blocked primers, which exhibited reduced extension efficiency in the absence of primer cleavage by RNase H2. We took the same principle of the match-specific cleavage of RNA: DNA hybrid pairing by RNase H2 for PROMER technology and developed it into more versatile tool capable of detecting rare and shorter targets in the mixture of genomic DNA with a high degree of precision. The blocking group, 3′-end DNA bases of the PROMER serves two functions: it quenches the fluorophore attached to the extension group and it creates a steric hindrance that prevents DNA polymerase II from elongating the strand if cleavage does not occur. PROMER technology only requires a single primer and a PROMER, while a conventional probe-based PCR (such as TaqMan™) needs an additional pair of primers along with the specific probe (Fig. 1B). If there is a mismatch in the cleavage group (e.g. wild-type allele with the mutant-matching PROMER), no cleavage occurs resulting in no amplification from the PROMER PCR (Fig. 1C). However, if the mutant allele is present, the PROMER PCR will generate real-time amplification (Fig. 1C).

Figure 1.

Figure 1.

Schematic representation of the PROMER technology. (A) Key components of the PROMER system. (B) PROMER PCR requires the use of one PROMER and one Primer for amplification. (C) Mechanism of mutant-specific amplification using PROMER technology. R represents reporter fluorescent dyes (FAM, HEX, or Cy5), while Q represents the quencher molecule.

Validation of PROMER technology using synthetic templates of KRAS mutations

To validate PROMER technology’s ability to detect point mutations accurately, we chose the human KRAS gene as our test subject. KRAS has various mutations at its 12th and 13th Glycine residues that require specific probes to differentiate them [42], which would determine the therapeutic regimen developed for the specific mutations. We synthesized total of 15 KRAS DNA templates including 13 known point mutations of G12C/S/R (at the 34th base), G12/V/D/A (at the 35th base), G13C/S/R (at the 37th base), G13/V/D/A (at the 38th base), Q61H (at the 183rd base), and two wild-types (Supplementary Table S1). We also created the PROMERs specific to these mutations, with selected fluorophore conjugated at its 5′-end and the quencher at its 3′-end (Supplementary Table S2). Serially diluted synthetic templates ranging from 1 × 105 copies to one copy were tested for the detection limit of each PROMER (Table 1). The PROMER PCR successfully detected one copy of the designated synthetic template for G12C/S/R/V/A, G13C/V/D, and Q61H (Table 1), while PROMERs for G12D and G13S/R/A detected 10 copies of the template (Table 1). There was no non-specific amplification in the no template control (NTC), indicating that the PROMER PCR can specifically detect low-copy number targets.

Table 1.

PROMER PCR detects low level of synthetic template with KRAS mutations.

Template copya Ct mean Template copya Ct mean
graphic file with name bpae041ilf1.jpg 1 × 105 22.756 graphic file with name bpae041ilf8.jpg 1 × 105 21.720
1 × 104 26.377 1 × 104 25.094
1 × 103 30.558 1 × 103 28.494
1 × 102 34.623 1 × 102 31.823
1 × 101 37.721 1 × 101 35.513
1 × 100 44.193 1 × 100 37.888
NTC ND NTC ND
graphic file with name bpae041ilf2.jpg 1 × 105 20.668 graphic file with name bpae041ilf9.jpg 1 × 105 21.401
1 × 104 23.779 1 × 104 25.562
1 × 103 28.108 1 × 103 29.796
1 × 102 30.961 1 × 102 33.184
1 × 101 36.509 1 × 101 37.076
1 × 100 36.087 1 × 100 ND
NTC ND NTC ND
graphic file with name bpae041ilf3.jpg 1 × 105 22.896 graphic file with name bpae041ilf10.jpg 1 × 105 24.101
1 × 104 26.941 1 × 104 27.697
1 × 103 30.319 1 × 103 32.622
1 × 102 34.751 1 × 102 36.273
1 × 101 37.854 1 × 101 41.067
1 × 100 40.883 1 × 100 ND
NTC ND NTC ND
graphic file with name bpae041ilf4.jpg 1 × 105 22.925 graphic file with name bpae041ilf11.jpg 1 × 105 20.571
1 × 104 26.942 1 × 104 23.565
1 × 103 30.255 1 × 103 27.439
1 × 102 35.160 1 × 102 29.888
1 × 101 37.028 1 × 101 35.469
1 × 100 39.158 1 × 100 35.328
NTC ND NTC ND
graphic file with name bpae041ilf5.jpg 1 × 105 22.004 graphic file with name bpae041ilf12.jpg 1 × 105 22.090
1 × 104 25.714 1 × 104 24.814
1 × 103 29.711 1 × 103 28.947
1 × 102 32.924 1 × 102 32.103
1 × 101 36.061 1 × 101 35.849
1 × 100 ND 1 × 100 37.582
NTC ND NTC ND
Inline graphic Inline graphic 1 × 105 22.980 graphic file with name bpae041ilf13.jpg 1 × 105 21.685
1 × 104 28.599 1 × 104 24.627
1 × 103 31.224 1 × 103 28.454
1 × 102 35.066 1 × 102 31.271
1 × 101 40.726 1 × 101 38.292
1 × 100 41.562 1 × 100 ND
NTC ND NTC ND
graphic file with name bpae041ilf7.jpg 1 × 105 22.018 #: number
NTC: No Template Control
ND: Not Detected
1 × 104 25.129
1 × 103 28.554
1 × 102 31.742
1 × 101 35.451
1 × 100 35.841
NTC ND

Each reaction was performed in triplicates.

a

Number.

ND, not detected.

Validation of PROMER technology using genomic DNA from human cancer cell lines with KRAS mutations

We conducted further validation of the PROMER technology to assess its ability to detect human genomic DNA with KRAS mutations and compared its efficacy to that of synthetic templates. Human cancer cell lines with KRAS mutations of G12C/S/R/V/D/A, G13C/D, and Q61H were obtained from ATCC (Supplementary Table S3) and their genomic DNAs were prepared for use as test templates. Serial dilutions of the genomic DNAs were tested using PROMER PCR, with copy numbers ranging from 1 × 104 to 1 × 10° for each genotype, as well as NTC (Supplementary Table S4). The Profit analysis was employed to evaluate the detectability of the PROMER PCR, with a detection probability of 93% or higher considered as a detectable copy number (Supplementary Table S5) [43]. The linearity between the tested copy numbers and the average Ct values was also determined. All tested PROMERS were able to detect one copy of each genomic DNA (Table 2 and Supplementary Table S6), with a coefficient of determination (R2) value of ≥0.99 DNA (Table 2 and Supplementary Table S7).

Table 2.

Detection limit and linearity of PROMER PCR for genomic DNAs with KRAS mutation.

Genotype Cell line Detection Limit (Copy number) R 2
G12C MIA PaCa-2 1 0.994
G12S A549 1 0.993
G12R MDA-MB-134VI 1 0.992
G12V SW620 1 0.999
G12D SNU-C2B 1 0.990
G12A SW1116 1 0.997
G13C NCI-H1734 1 0.996
G13D HCT-15 1 0.994
Q61H NCI-H460 1 0.994

Data used for the table are presented in Supplementary Table S5.

We also determined the detection specificity of PROMER PCR for KRAS mutations. Mutant genomic DNAs of 1 ×103, 1 × 102, 1 × 101, 1 × 100, and 0 copies in the presence of 1 × 104 copies of KRAS wild-type genomic DNA from NCI-H1975 cells were tested as templates. We compared the detection capability of PROMER PCR with commercial TaqMan assays for KRAS single-point mutation. Results showed that PROMER PCR exhibited comparable (G12C/S/A, G13C/D, and Q61H) or superior (G12S/R/V) sensitivity to the commercial probe-based PCR for the tested mutations (Table 3 and Supplementary Table S6).

Table 3.

Detection Specificity of PROMER and TaqMan PCR for genomic DNAs with KRAS mutation.

Genotype Cell line Detection limit (copy number) PROMER PCR Detection limit (copy number) TaqMan PCR
G12C MIA PaCa-2 1 1
G12S A549 10 10
G12R MDA-MB-134VI 1 NT
G12V SW620 1 10
G12D SNU-C2B 1 10
G12A SW1116 10 10
G13C NCI-H1734 1 1
G13D HCT-15 10 10
Q61H NCI-H460 1 1

All reactions were performed in the presence of 104 copies of the wild-type genomic DNA. Data used for the table are presented in Supplementary Table S6. NT, not tested.

We conducted a comprehensive examination of all KRAS G12 variants (G12C/S/R/V/D/A) to assess their cross-reactivity with other G12 types. We tested PROMERS for G12 variants (n = 20) against genomic DNAs of varying quantities (15, 30, 45, and 60 ng) prepared from cell lines representing each mutant type and the wild-type (Supplementary Table S3). All PROMERs demonstrated robust PCR amplification with the corresponding genomic DNA (Table 4). In contrast, when tested against non-matching genomic DNA, the PROMERs exhibited negligible PCR amplification (Table 4 and Supplementary Table S8). By setting a Ct cut-off value of 38, we observed no non-specific amplification for all G12 PROMERs. These results further confirm the specificity of our PROMER technology.

Table 4.

Detection Specificity of G12 PROMERs for genomic DNAs with other G12 mutations.

PROMER (Cell line) Target Templates
Template amount
G12C G12S G12R G12V G12D G12A
Wild-type (HEK293) 0/20 0/20 0/20 1/20 1/20 0/20 15 ng
0/20 2/20 0/20 0/20 0/20 0/20 30 ng
0/20 5/20 0/20 0/20 1/20 0/20 45 ng
0/20 1/20 0/20 0/20 3/20 0/20 60 ng
G12C (SW1573) 20/20 (28.50) 0/20 0/20 0/20 0/20 0/20 15 ng
20/20 (28.00) 0/20 3/20 0/20 0/20 0/20 30 ng
20/20 (27.63) 0/20 2/20 0/20 0/20 0/20 45 ng
20/20 (27.39) 0/20 1/20 0/20 0/20 0/20 60 ng
G12S (A549) 0/20 20/20 (27.55) 0/20 0/20 1/20 0/20 15 ng
0/20 20/20 (26.56) 0/20 0/20 0/20 0/20 30 ng
0/20 20/20 (25.96) 0/20 0/20 0/20 0/20 45 ng
0/20 20/20 (25.97) 0/20 0/20 0/20 0/20 60 ng
G12R (MDA-MB-134VI) 0/20 1/20 20/20 (29.68) 0/20 0/20 0/20 15 ng
0/20 0/20 20/20 (28.71) 0/20 0/20 0/20 30 ng
0/20 0/20 20/20 (28.03) 0/20 0/20 0/20 45 ng
0/20 1/20 20/20 (28.50) 0/20 0/20 0/20 60 ng
G12V (SW620) 0/20 0/20 0/20 20/20 (25.50) 0/20 0/20 15 ng
0/20 0/20 0/20 20/20 (24.69) 0/20 0/20 30 ng
0/20 0/20 0/20 20/20 (24.28) 1/20 0/20 45 ng
0/20 0/20 0/20 20/20 (24.03) 0/20 0/20 60 ng
G12D (SNU-C2B) 0/20 0/20 0/20 0/20 20/20 (29.88) 0/20 15 ng
0/20 0/20 0/20 0/20 20/20 (29.27) 0/20 30 ng
0/20 0/20 0/20 0/20 20/20 (29.04) 0/20 45 ng
0/20 0/20 0/20 0/20 20/20 (28.83) 0/20 60 ng
G12A (NCI-H2009) 0/20 0/20 0/20 1/20 1/20 20/20 (30.79) 15 ng
0/20 0/20 0/20 2/20 1/20 20/20 (30.24) 30 ng
0/20 1/20 0/20 6/20 0/20 20/20 (29.66) 45 ng
0/20 0/20 0/20 5/20 2/20 20/20 (28.95) 60 ng

Number represents frequency of amplifications per 20 repeated PCR reactions. Numbers in parenthesis represent the average Ct values for the matching genotypes. Ct values for all non-matching genotypes are greater than 38. Data used for the table are presented in Supplementary Table S8.

Multiplex detection of the PROMER PCR

Given the high specificity of the PROMER technology, which requires an exact match between the target template and the PROMER for PCR amplification, we hypothesized that PROMER PCR could enable multiplex PCR with mixture of the PRPMERs to detect target DNA in a single well. We designed three unique PROMERs with different fluorophores (HEX, FAM, and Cy5) for this application (Supplementary Table S2). The mixture for three PROMERs targeting KRAS G12C/S/R and G12V/D/A was tested for its ability to specifically detect its target using serially diluted genomic DNA from representative cell lines mixed with 1 × 105 copies of wild-type genomic DNA (Supplementary Table S9). All tested PROMER multiplex PCRs were capable of detecting the designated mutant genomic DNA (Table 5).

Table 5.

Multiplex PROMER PCR.

PROMER multiplex Template genotype Detection limit (copya) Template genotype Detection limit
G12C/S/R G12C 10 G12S 5
graphic file with name bpae041ilf14.jpg graphic file with name bpae041ilf18.jpg
G12R 1 Wild-type ND
graphic file with name bpae041ilf15.jpg graphic file with name bpae041ilf19.jpg
G12V/D/A G12V 1 G12D 1
graphic file with name bpae041ilf16.jpg graphic file with name bpae041ilf20.jpg
G12A 1 Wild-type ND
graphic file with name bpae041ilf17.jpg graphic file with name bpae041ilf21.jpg

Mixture for three PROMERs (G12C/S/R or G12V/D/A) were tested in the presence of 105 copies of wild-type genomic DNA. Each reaction was performed in triplicates. a: copy number. Data used for the table are presented in Supplementary Table S9.

Detection of ctDNA from plasma of tumor-bearing mice using the PROMER PCR

We further sought to examine the PROMER technology for its capability to detect ctDNA in blood plasma originating from the tumor. Subcutaneous (SC) tumors of varying sizes were developed using cancer cell lines for KRAS G12C/S/V/D/A, G13D, and Q61H (Supplementary Table S3) in NSG (NOD/Shi-scid/IL-2Rγnull) mice. MDA-MB-134VI (G12R) and NCI-H1734 (G13C) did not develop SC tumors in NSG mice. We isolated cfDNA from the peripheral blood of the animals and its quantity and average size of cfDNA was measured using Agilent 2100 Analyzer (Supplementary Fig. S1). The amount of the isolated cfDNA from each model varied but we observed a positive correlation between the tumor volume and the amount of isolated cfDNA (Fig. 2A and Supplementary Table S10). We successfully detected mutation-containing ctDNA from the cfDNA using the PROMER PCR and commercial KRAS detection agent purchased from ABI (Fig. 2B and Supplementary Table S10). The copy numbers were calculated using the standard curves generated with purified genomic DNA from each cell line (Supplementary Table S7). It is important to note that ctDNA quantity can vary depending on tumor type, size, and stage [23, 24, 27, 29]. The observed poor correlation between cfDNA copy number and detected ctDNA (Fig. 2B) may reflect the inherent heterogeneity of ctDNA. Nevertheless, both PROMER and ABI PCRs detected comparable copy numbers from each sample (Fig. 2C and Supplementary Table S10).

Figure 2.

Figure 2.

PROMER PCR detects ctDNA from plasma of mice with subcutaneous tumors with a KRAS mutation. (A) Comparison of tumor volumes and the amount of isolated cfDNA. (B) Relationship between the amount of isolated cfDNA and the copy number of detected ctDNA as measured by PROMER PCR and TaqMan PCR. (C) Correlation analysis between ctDNA copy numbers detected by PROMER and TaqMan PCR. Each data point represents a value obtained from a single mouse.

Discussion

In the era of precision medicine, the accurate identification and quantification of rare disease-originated nucleic acids from liquid biopsies have become increasingly important. These nucleic acids can provide valuable information for disease management, including the selection of treatment options, monitoring treatment efficacy, predicting prognosis, and identifying and characterizing acquired drug resistance [5]. However, detecting these disease-specific nucleic acids poses a significant challenge due to their rarity and subtle differences from their wild-type counterparts. High detection sensitivity and specificity are therefore required.

To address this challenge, we present the PROMER technology, a new real-time PCR method capable of precisely detecting rare nucleic acids. The specificity of PROMER technology comes from its unique design of PROMER and the matching sequence-specific cleavage of RNase H2 (Fig. 1). The generation of a PROMER, a DNA–RNA–DNA hybrid specific to its target, requires careful optimization of both its length and the positioning of the hybrid within the PROMER itself. In order to attain high levels of sensitivity and specificity, we performed further optimization in the PCR reaction in conjunction with sequence optimization of the PROMER (Supplementary Table S2). This involved fine-tuning parameters such as the concentration of the PROMER, the annealing temperature, and the extension temperature (Supplementary Table S4). Through our optimization process, we discovered that a higher temperature of 64°C for both annealing and extension steps yielded the highest specificity in detecting mutant templates (Supplementary Table S4). Utilization of a lower temperature for annealing and extension (below 60°C) could result in non-specific PCR reactions with the wild-type template (Supplementary Table S11). In order to facilitate the use of a higher annealing and extension temperature, our design objective for the PROMER was to achieve a melting temperature (TM) within the range of 61–62°C. Our validation data with synthetic templates and genomic DNA from human cancer cell lines with KRAS variant mutations demonstrate the efficiency of PROMER PCR (Tables 1 and 2). Further extensive testing of PROMER PCR with an excess amount of wild-type and genomic DNA of other variants (Table 4) confirms its detection specificity. Verifying the target-specific amplification by PROMER PCR through Sanger sequencing of the amplicons would provide more convincing evidence. However, due to the short length of the PROMER PCR amplicons, Sanger sequencing becomes impractical. This is because the sequencing reads for the first 40–50 base pairs (bp) are often unreliable, and the location of the mutation for our amplicon is within 39 bp from the end of the primer. Furthermore, the absence of PCR amplification of the mutant PROMER PCR with non-matching templates indirectly confirms the specificity of our mutation-specific PCR amplification.

PROMER technology employs a unique approach by using a single primer and a PROMER, which serves dual roles as both a probe and a primer (Fig. 1). Combining a primer and a real-time probe has been tested for PCR with high-fidelity polymerase for the purpose of detection of viral infection [44]. This feature facilitates the design and generation of shorter amplified PCR products compared to conventional real-time PCR methods that require two primers and a separate probe. The resulting shorter PCR amplicons offer several distinct advantages. One such advantage pertains to ctDNA, which exists in patient plasma as short double-stranded DNA fragments with an average size of 166 bp, corresponding to the size of DNA wrapped around a nucleosome [45]. Conventional real-time PCR methods may fail to detect variants located at either end of these ctDNA fragments. However, PROMER technology can detect its target regardless of the variant’s position within the short ctDNA. In addition, PROMER technology can be effectively employed for real-time PCR of both AT-rich and GC-rich templates. For AT-rich targets, the optimization of probe binding conditions poses a challenge, as the melting temperature (TM) should exceed that of the primers by at least 5°C. This limitation is circumvented in PROMER PCR, as the PROMER functions as both a probe and a primer. In the case of GC-rich templates, the introduction of additional mismatch base pairs 3′ to the cleavage group (Fig. 1A) can resolve issues arising from high TMs. Another significant advantage is that PROMER PCR can also detect short targets such as miRNA. Furthermore, the high detection specificity of PROMER technology enables precise differentiation of a miRNA from its isotypes, which often exhibit subtle sequence and structural variations. A potential concern when combining a primer and a probe for real-time PCR is the increased likelihood of non-specific amplification. This typically occurs through the formation of primer dimers or non-specific annealing of the PROMER. However, our PROMER technology is designed to address this issue. The cleavage of primer dimers by RNase H2 denatures the primer dimers when they form. Furthermore, through the careful optimization of the PROMER sequence and PCR conditions, as described earlier, we can significantly reduce the chances of non-specific annealing of the PROMER. We believe these measures effectively address the potential concerns related to non-specific amplification, thereby enhancing the reliability and accuracy of our PROMER technology. Previously, the 3TEC-PCR [46] adopted a similar approach, utilizing Tth endonuclease IV in place of RNase H2 to facilitate sequence-specific cleavage of its primer-probe hybrid. While 3TEC-PCR demonstrated impressive sensitivity, its capacity to detect lower copy numbers of its target amidst a significant quantity of its wild-type counterpart is yet to be investigated.

We selected KRAS mutations as our testing templates for the PROMER technology because they represent a challenging and clinically relevant scenario. KRAS mutations are involved in several cancers such as colorectal, pancreatic, non-small cell lung cancers, oesophageal adenocarcinoma/gastroesophageal junction cancer, invasive ductal carcinoma, stomach adenocarcinoma, and undifferentiated endometrial carcinoma [42]. Moreover, KRAS mutations can occur at multiple positions and types within the same gene, requiring high specificity of detection [47]. Furthermore, KRAS mutations can vary among different tumor sites or liquid biopsies from the same patient due to tumor heterogeneity [48]. These clinical situations demand a simple, multiplexed, reliable detection technology for rare nucleic acids from the liquid biopsy. PROMER technology has the potential to provide a solution for comprehensive KRAS variant detection. Our multiplexed testing results and in vivo examination of ctDNA from animal models of human KRAS-driven cancers support the capability of PROMER technology (Table 5 and Fig. 2).

In summary, we have developed a new real-time PCR method, the PROMER technology that can precisely detect and quantify rare nucleic acids from liquid biopsies. The PROMER technology has several advantages over conventional real-time PCR methods, such as shorter amplicons, higher specificity, and multiplexing capability. We have demonstrated the applicability of the PROMER technology for the detection and quantification of KRAS mutations, which are clinically relevant for many cancers. The PROMER technology can provide valuable information for disease management, such as treatment selection, monitoring, prognosis, and resistance. We believe that the PROMER technology is a promising tool for precision medicine in various diseases.

Supplementary Material

bpae041_Supplementary_Data

Contributor Information

Hwanhee Nam, Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD 21218, United States.

Esder Lee, NuriBio Co., Ltd, Anyang-si, Gyeonggi-Do, 14058, Republic of Korea.

Hichang Yang, NuriBio Co., Ltd, Anyang-si, Gyeonggi-Do, 14058, Republic of Korea.

Kyeyoon Lee, NuriBio Co., Ltd, Anyang-si, Gyeonggi-Do, 14058, Republic of Korea.

Taeho Kwak, NuriBio Co., Ltd, Anyang-si, Gyeonggi-Do, 14058, Republic of Korea.

Dain Kim, NuriBio Co., Ltd, Anyang-si, Gyeonggi-Do, 14058, Republic of Korea.

Hyemin Kim, NuriBio Co., Ltd, Anyang-si, Gyeonggi-Do, 14058, Republic of Korea.

Mihwa Yang, NuriBio Co., Ltd, Anyang-si, Gyeonggi-Do, 14058, Republic of Korea.

Younjoo Yang, NuriBio Co., Ltd, Anyang-si, Gyeonggi-Do, 14058, Republic of Korea.

Seungwan Son, NuriBio Co., Ltd, Anyang-si, Gyeonggi-Do, 14058, Republic of Korea.

Young-Hyean Nam, NuriBio Co., Ltd, Anyang-si, Gyeonggi-Do, 14058, Republic of Korea.

Il Minn, Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD 21218, United States; Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins Medical Institutions, Baltimore, MD 21287, United States.

Author contributions

Hwanhee Nam (Conceptualization [supporting], Data curation [supporting], Formal analysis [supporting], Investigation [supporting], Methodology [supporting], Validation [supporting], Writing—original draft [supporting], and Writing—review & editing [supporting]), Esder Lee (Data curation [supporting], Formal analysis [supporting], Methodology [supporting], and Writing—review & editing [supporting]), Hichang Yang (Data curation [supporting], Formal analysis [supporting], Methodology [supporting], and Writing—review & editing [supporting]), Kyeyoon Lee (Data curation [supporting], Formal analysis [supporting], Methodology [supporting], and Writing—review & editing [supporting]), Taeho Kwak (Data curation [supporting], Formal analysis [supporting], Methodology [supporting], and Writing—review & editing [supporting]), Dain Kim (Data curation [supporting], Formal analysis [supporting], Methodology [supporting], and Writing—review & editing [supporting]), Hyemin Kim (Data curation [supporting], Formal analysis [supporting], Methodology [supporting], and Writing—review & editing [supporting]), Mihwa Yang (Data curation [supporting], Formal analysis [supporting], Methodology [supporting], and Writing—review & editing [supporting]), Younjoo Yang (Data curation [supporting], Formal analysis [supporting], Methodology [supporting], and Writing—review & editing [supporting]), Seungwan Son (Data curation [supporting], Formal analysis [supporting], Methodology [supporting], and Writing—review & editing [supporting]), Young-Hyean Nam (Conceptualization [equal], Data curation [supporting], Formal analysis [supporting], Investigation [supporting], Methodology [equal], Project administration [equal], Resources [equal], Supervision [equal], Validation [equal], Writing—original draft [supporting], and Writing—review & editing [supporting]), and IL MINN (Conceptualization [equal], Data curation [lead], Formal analysis [lead], Funding acquisition [lead], Investigation [lead], Methodology [equal], Project administration [equal], Resources [equal], Supervision [equal], Validation [equal], Writing—original draft [lead], and Writing—review & editing [lead])

Supplementary data

Supplementary data is available at Biology Methods & Protocols online.

Conflict of interest statement. E.L., H.Y., K.L., T.K., D.K., H.K., M.Y., Y.Y., S.S., and Y.N. are employees of Nuribio Co., Ltd. Y.N. is an inventor and Nuribio Co., Ltd is an assignee of the patent WO2017142297A1 (PCT/KR2017/001632) [38].

Funding

This work was supported by a funding from Nuribio Co., Ltd through a contract research agreement.

Data availability

The data underlying this article are available in the article and in its Online Supplementary material.

References

  • 1. Alix-Panabieres C, Pantel K.. Liquid biopsy: from discovery to clinical application. Cancer Discov 2021;11:858–73. [DOI] [PubMed] [Google Scholar]
  • 2. Mandel P, Metais P.. [Nuclear acids in human blood plasma]. C R Seances Soc Biol Fil 1948;142:241–3. [PubMed] [Google Scholar]
  • 3. Leon SA, Shapiro B, Sklaroff DM. et al. Free DNA in the serum of cancer patients and the effect of therapy. Cancer Res 1977;37:646–50. [PubMed] [Google Scholar]
  • 4. Lennon AM, Buchanan AH, Kinde I. et al. Feasibility of blood testing combined with PET-CT to screen for cancer and guide intervention. Science 2020;369: [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5. Cescon DW, Bratman SV, Chan SM. et al. Circulating tumor DNA and liquid biopsy in oncology. Nat Cancer 2020;1:276–90. [DOI] [PubMed] [Google Scholar]
  • 6. Chabon JJ, Hamilton EG, Kurtz DM. et al. Integrating genomic features for non-invasive early lung cancer detection. Nature 2020;580:245–51. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7. Chen X, Gole J, Gore A. et al. Non-invasive early detection of cancer four years before conventional diagnosis using a blood test. Nat Commun 2020;11:3475. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8. Weber S, van der Leest P, Donker HC. et al. Dynamic changes of circulating tumor DNA predict clinical outcome in patients with advanced non-small-cell lung cancer treated with immune checkpoint inhibitors. JCO Precis Oncol 2021;5:1540–53. [DOI] [PubMed] [Google Scholar]
  • 9. Zhou Q, Gampenrieder SP, Frantal S. et al. Persistence of ctDNA in patients with breast cancer during neoadjuvant treatment is a significant predictor of poor tumor response. Clin Cancer Res 2022;28:697–707. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10. Pellini B, Chaudhuri AA.. Circulating tumor DNA minimal residual disease detection of non-small-cell lung cancer treated with curative intent. J Clin Oncol 2022;40:567–75. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11. Abbosh C, Birkbak NJ, Wilson GA; PEACE Consortium. Phylogenetic ctDNA analysis depicts early-stage lung cancer evolution. Nature 2017;545:446–51. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12. Garcia-Murillas I, Chopra N, Comino-Méndez I. et al. Assessment of molecular relapse detection in early-stage breast cancer. JAMA Oncol 2019;5:1473–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13. Azad TD, Chaudhuri AA, Fang P. et al. Circulating tumor DNA analysis for detection of minimal residual disease after chemoradiotherapy for localized esophageal cancer. Gastroenterology 2020;158:494–505 e496. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14. Turner NC, Kingston B, Kilburn LS. et al. Circulating tumour DNA analysis to direct therapy in advanced breast cancer (plasmaMATCH): a multicentre, multicohort, phase 2a, platform trial. Lancet Oncol 2020;21:1296–308. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15. Sicklick JK, Kato S, Okamura R. et al. Molecular profiling of cancer patients enables personalized combination therapy: the I-PREDICT study. Nat Med 2019;25:744–50. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16. Jacob S, Davis AA, Gerratana L. et al. The use of serial circulating tumor DNA to detect resistance alterations in progressive metastatic breast cancer. Clin Cancer Res 2021;27:1361–70. [DOI] [PubMed] [Google Scholar]
  • 17. Chabon JJ, Simmons AD, Lovejoy AF. et al. Circulating tumour DNA profiling reveals heterogeneity of EGFR inhibitor resistance mechanisms in lung cancer patients. Nat Commun 2016;7:11815. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18. Heitzer E, van den Broek D, Denis MG. et al. Recommendations for a practical implementation of circulating tumor DNA mutation testing in metastatic non-small-cell lung cancer. ESMO Open 2022;7:100399. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19. Finan C, Gaulton A, Kruger FA. et al. The druggable genome and support for target identification and validation in drug development. Sci Transl Med 2017;9:eaag1166. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20. Bayle A, Belcaid L, Aldea M. et al. Clinical utility of circulating tumor DNA sequencing with a large panel: a National Center for Precision Medicine (PRISM) study. Ann Oncol 2023;34:389–96. [DOI] [PubMed] [Google Scholar]
  • 21. Newman AM, Bratman SV, To J. et al. An ultrasensitive method for quantitating circulating tumor DNA with broad patient coverage. Nat Med 2014;20:548–54. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22. Newman AM, Lovejoy AF, Klass DM. et al. Integrated digital error suppression for improved detection of circulating tumor DNA. Nat Biotechnol 2016;34:547–55. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23. Diehl F, Schmidt K, Choti MA. et al. Circulating mutant DNA to assess tumor dynamics. Nat Med 2008;14:985–90. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24. Bettegowda C, Sausen M, Leary RJ. et al. Detection of circulating tumor DNA in early- and late-stage human malignancies. Sci Transl Med 2014;6:224ra24. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25. Lui YY, Chik KW, Chiu RW. et al. Predominant hematopoietic origin of cell-free DNA in plasma and serum after sex-mismatched bone marrow transplantation. Clin Chem 2002;48:421–7. [PubMed] [Google Scholar]
  • 26. Zheng YWL, Chan KCA, Sun H. et al. Nonhematopoietically derived DNA is shorter than hematopoietically derived DNA in plasma: a transplantation model. Clin Chem 2012;58:549–58. [DOI] [PubMed] [Google Scholar]
  • 27. Ramalingam N, Jeffrey SS.. Future of liquid biopsies with growing technological and bioinformatics studies: opportunities and challenges in discovering tumor heterogeneity with single-cell level analysis. Cancer J 2018;24:104–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28. Reiter JG, Baretti M, Gerold JM. et al. An analysis of genetic heterogeneity in untreated cancers. Nat Rev Cancer 2019;19:639–50. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29. Corcoran RB, Chabner BA.. Application of cell-free DNA analysis to cancer treatment. N Engl J Med 2018;379:1754–65. [DOI] [PubMed] [Google Scholar]
  • 30. Heitzer E, Haque IS, Roberts CES. et al. Current and future perspectives of liquid biopsies in genomics-driven oncology. Nat Rev Genet 2019;20:71–88. [DOI] [PubMed] [Google Scholar]
  • 31. Murtaza M, Dawson SJ, Tsui DWY. et al. Non-invasive analysis of acquired resistance to cancer therapy by sequencing of plasma DNA. Nature 2013;497:108–12. [DOI] [PubMed] [Google Scholar]
  • 32. Forshew T, Murtaza M, Parkinson C. et al. Noninvasive identification and monitoring of cancer mutations by targeted deep sequencing of plasma DNA. Sci Transl Med 2012;4:136ra68. [DOI] [PubMed] [Google Scholar]
  • 33. Kinde I, Wu J, Papadopoulos N. et al. Detection and quantification of rare mutations with massively parallel sequencing. Proc Natl Acad Sci USA 2011;108:9530–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34. Stahlberg A, Krzyzanowski PM, Egyud M. et al. Simple multiplexed PCR-based barcoding of DNA for ultrasensitive mutation detection by next-generation sequencing. Nat Protoc 2017;12:664–82. [DOI] [PubMed] [Google Scholar]
  • 35. Perkins G, Lu H, Garlan F. et al. Droplet-based digital PCR: application in cancer research. Adv Clin Chem 2017;79:43–91. [DOI] [PubMed] [Google Scholar]
  • 36. Suzuki Y, Sekiya T, Hayashi K.. Allele-specific polymerase chain reaction: a method for amplification and sequence determination of a single component among a mixture of sequence variants. Anal Biochem 1991;192:82–4. [DOI] [PubMed] [Google Scholar]
  • 37. Li J, Wang L, Mamon H. et al. Replacing PCR with COLD-PCR enriches variant DNA sequences and redefines the sensitivity of genetic testing. Nat Med 2008;14:579–84. [DOI] [PubMed] [Google Scholar]
  • 38. Nam YH. Single-stranded nucleic acid for detecting nucleic acid or protein in real time and detection method using same. Patent number: WO2017142297A1 (2017). [Google Scholar]
  • 39. Le Laz S, Le Goaziou A, Henneke G.. Structure-specific nuclease activities of Pyrococcus abyssi RNase HII. J Bacteriol 2010;192:3689–98. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40. Kitamura S, Fujishima K, Sato A. et al. Characterization of RNase HII substrate recognition using RNase HII-argonaute chimaeric enzymes from Pyrococcus furiosus. Biochem J 2010;426:337–44. [DOI] [PubMed] [Google Scholar]
  • 41. Dobosy JR, Rose SD, Beltz KR. et al. RNase H-dependent PCR (rhPCR): improved specificity and single nucleotide polymorphism detection using blocked cleavable primers. BMC Biotechnol 2011;11:80. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42. Hofmann MH, Gerlach D, Misale S. et al. Expanding the reach of precision oncology by drugging all KRAS mutants. Cancer Discov 2022;12:924–37. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43. Kralik P, Ricchi M.. A basic guide to real time PCR in microbial diagnostics: definitions, parameters, and everything. Front Microbiol 2017;8:108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44. Zhang M, Liu K, Hu Y. et al. A novel quantitative PCR mediated by high-fidelity DNA polymerase. Sci Rep 2017;7:10365. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45. Diehl F, Li M, Dressman D. et al. Detection and quantification of mutations in the plasma of patients with colorectal tumors. Proc Natl Acad Sci USA 2005;102:16368–73. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46. Higgins O, Smith TJ.. 3' Tth endonuclease cleavage polymerase chain reaction (3TEC-PCR) technology for single-base-specific multiplex pathogen detection using a two-oligonucleotide system. Int J Mol Sci 2021;22:6061. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47. Macedo MP, Andrade LDB, Coudry R. et al. Multiple mutations in the Kras gene in colorectal cancer: review of the literature with two case reports. Int J Colorectal Dis 2011;26:1241–8. [DOI] [PubMed] [Google Scholar]
  • 48. Kulemann B, Rösch S, Seifert S. et al. Pancreatic cancer: circulating tumor cells and primary tumors show heterogeneous KRAS mutations. Sci Rep 2017;7:4510. [DOI] [PMC free article] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

bpae041_Supplementary_Data

Data Availability Statement

The data underlying this article are available in the article and in its Online Supplementary material.


Articles from Biology Methods & Protocols are provided here courtesy of Oxford University Press

RESOURCES