Somatic Mutations
Georgeta N Basturea (gbasturea at gmail dot com )
University of Miami, Florida, United States
DOI
//dx.doi.org/10.13070/mm.en.8.2673
Date
last modified : 2022-11-19; original version : 2018-11-30
Cite as
MATER METHODS 2018;8:2673

Somatic mutations are mutations acquired by non-germline cells and cannot be inherited by the offspring of the parent organism of the mutated cell, with the exception of, for example, canine transmissible venereal tumor [6]. Somatic mutations are important in the diversity of the antibodies, T cell receptors, and B cell receptors. They are frequently caused by environmental factors and accumulate in the DNA of any organism despite proficient DNA repair mechanisms. Somatic mutations are present in healthy tissues at a frequency of about 2-6 mutations per million bases [7], and about three somatic mutations per healthy human individual [8]. As a consequence, somatic cells present different genotypes within the same individual, a widespread phenomenon in healthy development and aging, known as somatic mosaicism [9, 10]. Somatic mutations accumulate during the aging process. Endogenous somatic mutations are a contributing factor in aging, since the rate of somatic mutations across mammlian species displays an inverse relationship with species lifespan [11]. K Nanki et al measured 0.019 single nucleotide variant per Mb per year among colonic epithelia [12]. Zhang L et al, using Single-Cell Multiple Displacement Amplification whole genome sequencing, found that the number of somatic mutations in human B lymphocytes increased from <500 per cell in newborns to >3,000 per cell in centenarians [13]. K Yoshida et al estimated 22 single-base substitutions per cell per year among the bronchial epithelial cells [14].

The implications of mosaicism in disease are not completely understood [15, 16], but recent studies suggest that it influences cancer [17-21], neurological and neuropsychiatric disorders [16, 22-25], ulcerative colitis [12], and various monogenic disorders [26]. In cancer cells, such mutations were found at high frequency. In a review by Martincorena and Campbell (2015) [18] it is stated that between 1000 and 20,000 point mutations and hundreds of insertions, deletions and rearrangements can be detected in cancers. These numbers were determined based on studies on millions of mutations in various cancer types [27-30]. COSMIC (the Catalogue Of Somatic Mutations In Cancer) [31] version 91, released in April 2020, includes almost 34,657,730 coding mutations across 1,443,198 samples, curated from 27,496 papers . The catalog is a great resource for cancer research, for example in [32, 33]. TCGA is another resource with somatic mutations in various cancers [32]. Somatic mutations on ENL likely contribute to Wilms tumour machanistically [34]. Researchers have started treating patients based on somatically mutated genes, for instance, a patient has been successfully treated with tumor-infiltrating lymphocytes reactive against mutant SLC3A2, KIAA0368, CADPS2 and CTSB [35].

Somatic mutations are not reduced to point mutations but can be any genomic variation: repeats, deletions, insertions, multiplication, loss of copy number, and others. Chromosomal somatic mutations occur when somatic cells divide. During this time, structural aberrations result from chromosome breakages and incorrect repairing or the unequal exchange of material during chromosome separation. Structural aberrations include deletions, when a part of the chromosome is missing; duplications when a portion of the chromosome appears twice; translocation when genetic material has been interchanged between non-homologous chromosomes or inversions, when part of the chromosome is in inverse orientations. Since such changes occur only in some cells, chromosomal mosaicism is observed.

One common concern when trying to identify and characterize somatic mutations is their low abundance. Due to the low frequency, somatic mutations are difficult to detect in bulk tissue samples, and amplification of template DNA is necessary before analysis. However, DNA amplification is an error-prone process, and real mutations may be hidden among these errors. Even the modern sequencing technologies fail to detect many somatic variants, especially the ones present in very few or single copies. The sequencing errors are high enough to mask real variations [36]. Sensitive methods are necessary for the detection and relative quantification of somatic mutations in biological material.

Due to the variety of mutation types and the difficulties mentioned above in detecting somatic mutations, a wide range of techniques have been and continue to be developed. PCR, electrophoretic, chromatography and sequencing methods are most commonly used for detecting either known (for diagnosis) or unknown point mutations. To these, probing methods are added for the detection of chromosomal aberrations. Detection methods can be specific to targeted chromosomal regions or used for whole exome, for example, in the study of canine transmissible venereal tumor [6] or colonic epithelium associated with ulcerative colitis []31853059, whole genome or transcriptome analysis.

Here we present classical and modern methods for identification of somatic variations. Each method is briefly described, and studies that successfully used it are exemplified. The list includes commonly used techniques, without being considered exhaustive. As new technologies are being developed, they are added to this collection. Table 1 provides a summary discussing usefulness, sensitivity and some advantages and disadvantages.

Method Advantage/Disadvantage Known/Unknown mutation Example studies
CRISPR-SNP-Chipa label-free technology based on a graphene field-effect transistor; unamplified genomic DNA samplesknown [37]
Amplification refractory mutation system (ARMS) and variantsRapid, easy to use, efficient, non-radioactive, semi-quantitative, selectivity (1 in 102 to 1 in 105) [38, 39] known [40, 41]
Real-time PCR (qPCR)Rapid, efficient, non-radioactive, quantitative but standard curve and amplification efficiency dependentknown [42, 43]
Reverse-transcription PCR (RT-PCR)Rapid, efficient, non-radioactive, applied to the study of mutations at the RNA level and chromosomal translocationsboth [44, 45]
Digital PCR (dPCR)Rapid, efficient, quantitative, amplification efficiency independent, non-radioactiveknown [46, 47]
Fluorescent-amplicon generation (FLAG) assayEfficient, non-radioactive, semi-quantitative, selectivity: 1 in 103 [2] known [2, 48]
Peptide-nucleic acids PCR (PNA-PCR)Non-radioactive, semi-quantitative, selectivity: 1 in 5x105 [49] ; require time for optimizationknown [50, 51]
Locked nucleic acids PCR (LNA-PCR)Non-radioactive, semi-quantitative, selectivity: 1 in 105 [52] ; require time for optimizationknown [53, 54]
Co-amplification at lower denaturation temperature PCR (COLD-PCR)Rapid, simple, selectivity: 1 in 104 [55], must know the precise denaturation temperature, applies only to short duplexes, limited by polymerase-induced errorsboth [56, 57]
Single–strand conformation polymorphism (SSCP)Simple, non-radioactive, screening, optimal fragment size 150-200 bp, detects 80-90% point mutations [58, 59] both [60]
Denaturing gradient gel electrophoresis (DGCE)Screening, good for small DNA fragments, almost 100% mutations can be detected [61] both [62, 63]
Constant denaturant capillary electrophoresis (CDCE)Rapid, good for screening small DNA fragments, selectivity: 1 in 103 [64] to 1 in 106 [65] both [64, 66]
Denaturing high-pressure liquid chromatography (dHPLC)Screening method, DNA fragments up to 1.5 kb [67], fast, can detect more than 90% point mutations [67, 68] both [69]
Mismatch Repair Detection (MRD)Screening method, works with large sample size and DNA fragments, can be laboriousboth [70]
Restriction-endonuclease based PCR (REMS-PCR)Efficient, non-radioactive, semi-quantitative, selectivity, selectivity: 1 in 2.5 x103 [71] known [48, 71]
High resolution melting (HRM)Fast, high-throughput scanning, non-radioactive, high sensitivity [72] and precision [73] both [72, 73]
Mass-ARRAYPermits analysis of multiple mutations and genes simultaneously, high sensitivityboth [74, 75]
DNA microarraysPermits analysis of multiple mutations and genes simultaneously, high sensitivityboth [76]
Multiplex ligation-dependent probe amplification (MLPA)Good for detecting gene deletions or duplications, can analyze multiple genes and multiple samples simultaneously [77, 78] both [78, 79]
DNA and RNA in-situ hybridization (DISH and RISH)Ideal for detecting gene deletions, duplications, or rearrangements;both [45, 80]
Direct sequencingSimple, accurate, preferred for point mutations screening in localized genesboth [81, 82]
Next-generation sequencing (NGS)Used for random DNA fragments, full genome, exosome or specific gene sequencingboth [11, 14]
Single-molecule real-time sequencing (SMTR sequencing)Sample enrichment, either DNA-seq or RNA-seqboth [45, 83]
Single-cell sequencingSample enrichment, either DNA-seq or RNA-seqboth [84, 85]
Table 1. Molecular methods for mutation detection.
PCR-based Methods for Detecting Known Small Mutations

Variations of PCR have been used to identify known small mutations.

Conventional PCR based detection of a single point or small insertion/deletion mutations.

Single point or small insertion/deletion mutations are commonly detected by the Amplification Refractory Mutation System (ARMS) technique and its variations. This method, also know as allele-specific PCR (ASP) or PCR amplification of specific alleles (PASA), is a conventional PCR-based method used to detect single base mutations in a complex pool of DNA molecules. In this method, mutations at known locations in the target sequences can be identified by performing two complementary PCR reactions with one common 5’- primer specific to the target DNA region, and two 3’- primers that are allele-specific. The only difference between these two primers is at a given base, such that one complements the wild-type sequence and the other the mutant. If the sample is homozygous amplification will only occur in one of the tubes; if the sample is heterozygous amplification will be seen in both tubes (Figure 1A). One essential factor in this method is the thermostable polymerase that lacks the 3’-5’ exonuclease activity, like Taq polymerase, used to avoid the elimination of the mutation in the mutant-specific primer. Agarose gel electrophoresis followed by ethidium bromide staining is commonly used to visualize the PCR products. This is a rapid and efficient method when the PCR conditions are properly chosen [38, 86, 87] and can also be used for amplification of target sequences containing small insertion or deletions.

Fluorescent labeling and DNA sequence analysis improve the technique, allowing for co-amplification of normal and mutant DNA fragments with different size primers [88, 89]. Relative quantification of allele proportions is possible by combining ARMS with analysis of the DNA product melting-point temperatures obtained from post-PCR fluorescent dissociation curves (DCA) [42, 90, 91]. A further improvement of the technique is seen in tetra-primer ARMS (T-ARMS) in which two common primers are used to amplify a DNA sequence as an internal control, while opposite orientation allele-specific primers help amplify size –specific fragments (Figure 1B) [40, 92-94]. Other variants of ARMS include competitive oligonucleotide priming (COP) [95], mutant enrichment PCR (enriched-EPCR or mutant-enriched - ME-PCR) [96, 97], mismatch amplification mutation assay (MAMA) [98], mutant allele-specific amplification (MASA) [99], and allele-specific competitive blocker-polymerase chain reaction (ACB-PCR) [100].

Somatic Mutations  figure 1
Figure 1. Conventional PCR based allele-specific amplification and mutant DNA detection. A) Steps of ARMS amplification of DNA at a known location; B) Tetra-primer ARMS variant technique.
Real-time PCR (qPCR)

A combination of allele-specific PCR and real-time PCR can also be used to detect minority alleles [42, 43, 101-104]. In real-time PCR the amount of PCR product is measured after each round of amplification using a fluorescent readout [105]. The amount of target sequence is determined based on a standard curve obtained from samples of known copy number, assuming that the amplification efficiencies of the sample and the standards are equivalent.

Reverse-transcription PCR (RT-PCR)

As in real-time PCR, the amplified material in reverse-transcription PCR is mRNA. In this technique, the mRNA in fist transcribed into a complementary DNA strand, which is subsequently amplified in a classical PCR amplification cascade. The final PCR product is sequenced by Sanger sequencing. This method proved useful in detecting unexpected RNA splice variants that contained various mutations (intra-exonic junctions, insertions, deletions, single nucleotide variations) in a recent study on small populations of neurons from Alzheimer’s disease patients [45]. In combination with other observations (see below), this finding led to the important conclusion that neuronal gene recombination may lead to the presence and the expression of short variants of normal genes, which might be a mechanism with significant consequences for the normal functioning and occurrence of diseases in the human brain.

This technique is ideal for the study of somatic mutation at the level of RNA as well as for detection of chromosomal translocations [44, 45, 106].

Digital PCR (dPCR)

In digital PCR the sample is diluted such that every dilution contains a minimum amount of target sequence, ideally one or none. Each dilution is used as a template for a PCR reaction, and the amplified product is detected by fluorescence. The distribution of target sequences in the diluted samples can be approximately determined by a Poisson’s distribution, while their concentration can be calculated based on the ratio of samples with positive amplification to total dilution samples. In dPCR, the fluorescent signal is measured at the end of the amplification and does not rely on comparisons with a standard curve. This way, the measurement is not dependent on the efficiency of the amplification reaction, as is the case in qPCR. Multiple somatic mutations have been identified as causes for several malformative disorders [107, 108], by using this method.

A comparison of the conventional, real-time and digital PCR techniques is shown in Figure 2.

Somatic Mutations  figure 2
Figure 2. Comparison of PCR-based techniques (from Quan et al, 2018 [1] ).
PCR with physically altered DNA (PNA-PCR and LNA-PCR)

Other variations of the PCR technique use modified nucleic acids, like peptide nucleic acids (PNAs) [50, 109, 110] or locked nucleic acids (LNAs) [53, 54, 111], as a substitute for primers. In these techniques, the wild-type DNA amplification is suppressed, and the mutant template is being amplified. Similarly, a DNA tag containing a highly thermostable restriction nuclease recognition site has been used in FLAG (fluorescent amplicon generation) assays that combine PNA probes and restriction endonuclease-mediated selective (REMS) PCR (Figure 3) [2, 48]

Somatic Mutations  figure 3
Figure 3. FLAG assay and PNA-PCR based selection of mutant templates. (Modified from Amicarelli et al, 2007 [2]. A) FLAG assay principle. A restriction nuclease sensitive site (PspGI) is inserted by DNA amplification only when the DNA sequence is wild-type. One of the primers is labeled with both a quencher (Q) and a fluorophore (F). When wild-type DNA is amplified, digestion leads to a fluorescent signal increase due to loss of quenching. B). PNA-mediated FLAG assay. When a PNA specific to a particular mutation is added to the reaction, the formed PNA/DNA hybrid inhibits primer annealing and DNA amplification such that no fluorescent signal is obtained. However, primer hybridization is not inhibited by incomplete complementarity of the PNA (single mismatch) leading to generation of a fluorescent signal. By conducting four distinct FLAG reactions with PNA probes specific for different mutations (GTT, GAT, GCT, TGT), the identity of the mutation can be inferred from the intensity of the fluorescent signals obtained.
Co-amplification at lower denaturation temperature PCR (COLD-PCR)

The COLD-PCR techniques is a single-step technique that permits the preferential amplification of DNA fragments with known or unknown mutations, based on the lower annealing temperature of mismatch-containing duplexes. Mutated DNA sequences are enriched during PCR by lowering the denaturing temperature such that imperfect pairs are denatured but not the fully complementary wild-type duplexes. A new round of primer annealing now has mostly mutant DNA available for pairing and replication [55, 112, 113]. Two forms of the technique have been developed. One permits the enrichment of all mutants (Full COLD-PCR – Figure 4 A) [114, 115], while the second only allows the amplification of melting temperature-reducing mutations (Fast COLD-PCR - Figure 4B) [116, 117].

Somatic Mutations  figure 4
Figure 4. Principle of COLD-PCR. A) Full COLD-PCR; B) Fast COLD-PCR.
Post-PCR methods for isolation of DNA with unknown mutations
Electrophoretic methods
Single–strand conformation polymorphism (SSCP)

This technique allows for the differentiation of wild-type and mutant DNA strands based on their mobility shifts produced by their different conformations when single-stranded. The separation of differently structured denatured DNA strands can be done either in gel [118] or through capillaries [119, 120], when an electric current is applied (GE- gel electrophoresis or CE – capillary electrophoresis SSCP). Figure 5 illustrates the principle of this technique.

Somatic Mutations  figure 5
Figure 5. Single-strand conformation polymorphism detection by gel electrophoresis (A), and capillary electrophoresis (B). In capillary electrophoreses fluorescently labeled DNA fragments are denatured, separated according to their migration, and detected based on their fluorescent signal.
Denaturing gradient gel electrophoresis (DGGE)

Denaturing gradient gel electrophoresis (DGGE) allows the screening of PCR-amplified DNA for single base mutations. In this technique, a mixture of DNA fragments of different sequences is separated by electrophoresis in an acrylamide gel containing a linearly increasing gradient of denaturant. More stable DNA fragments like the G-C rich fragments migrate faster, while denatured molecules move slower in the gel. In this manner, DNA fragments can be separated in the acrylamide gel. DNA fragments can be extracted from the gel, amplified and sequenced [61]. Tumor-specific mutations have been detected successfully by this method [121].

Constant denaturant capillary electrophoresis (CDCE)

CDCE is another technique that uses the power of denaturants, thermal or chemical, to separate different sequence DNA fragments through capillaries under electric currents. The method can be used to rapidly detect point mutations in candidate disease genes [122-125].

Chromatography-based methods

Separation of nucleic acids based on their size can be easily achieved by chromatography. In addition, when denaturants are added, the molecules can be separated based on their structure.

Denaturing high-pressure liquid chromatography (dHPLC)

Similarly to the other techniques that use denaturants to detect conformational changes in DNA molecules with a slightly different sequence, liquid chromatography can be used for separating such molecules based in differences in their column retention time [67, 126-130].

Enzyme-based methods

Enzymatic mutation detection takes advantage of natural processes in which DNA-cutting enzymes are involved.

Mismatch repair detection (MRD)

Several methods for detecting single nucleotide changes in DNA exploit the ability of the mismatch repair proteins to bind mismatched nucleotide pairs. One of the commonly used enzymes, the E. coli MutS can bind heteroduplexes with up to four mismatched pairs. The enzyme has been immobilized on a variety of solid supports (nylon, nitrocellulose, PVDF membrane) [131, 132] or to protein chip matrices [133, 134] and used to scan amplified DNA for fragments containing mismatches in vitro.

Moreover, in vivo detection of mismatch-containing duplexes has been developed. Faham and Cox (1995) [135] used E. coli to screen DNA fragments for variations. They cloned DNA fragments into two MRD plasmids of which one can, and one cannot express lacZ due to a 5bp insertion. They amplified the plasmids into both methylation deficient and wild-type bacterial strains. Extracted methylated and unmethylated plasmids were mixed, denatured, renatured, and subjected to digestion with specific restriction endonucleases. Since only fully methylated or unmethylated DNA is degraded, hemimethylated DNA is selected. This heteroduplexed plasmid is transformed in E. coli where it triggers the mismatch repair response, resulting in a repaired lacZ gene. A white colony grows and can be selected. In the absence of a mismatch, DNA replication produces both lacZ variants, and blue colonies grow. The method does not allow for the identification of the particular variations but can find all DNA fragments that contain mutations, and it can be adapted for scanning large genomic regions [70, 136-138].

Restriction endonuclease-based methods (REMS-PCR)

Mutant enrichment has been achieved in restriction endonuclease-mediated selective PCR (REMS-PCR) with the help of heat-resistant restriction enzymes that selectively destroy wild-type [2, 71, 104, 139] or mutant [140] DNA during PCR. A combination of restriction enzyme digestion and real-time PCR (real-time digestion PCR) proved to be efficient in detecting somatic mutations [141] Alterations in restriction enzymes’ recognition or cut sites as a result of point mutations can also be identified as a result of altered restriction digestion patterns in restriction fragment length polymorphism (RFPL) [142-145].

High-resolution melting (HRM)

High-resolution melting (HRM) curve analysis represents a fast, post-PCR high-throughput method for scanning somatic sequence alterations in target genes. In this method, the fluorescent signal from intercalating dyes in dsDNA is read as a measure of the degree of hybridization. dsDNA is denatured gradually by increasing temperature, and the decrease of the fluorescent signal is measured generating “melting” curves. Shifts of the melting curves permit the identification of mutant DNA fragments (Figure 6). The method has been successfully used to detect somatic mutations in multiple types of cancer [72, 146-149].

Somatic Mutations  figure 6
Figure 6. High-resolution melting (HRM) curve analysis of wild type and mutant DNA. A shift of the melting curve indicates a mutant DNA fragment.
Mass ARRAY technology

A novel technology developed by Sequenom (www.sequenom.com) - the Mass ARRAY system - combines PCR amplification, single-base primer extension and mass spectroscopy of DNA for mutation detection. Wild type and mutant DNA is amplified by multiplex PCR, and the primer extension reaction is performed with mass-modified terminator nucleotides. The product is analyzed, and mutant and wild type fragments are identified by MALDI-TOF (Matrix-Assisted Laser Desorption/Ionization – Time of Flight) [74, 75, 150].

Probing-based Methods
DNA microarrays

DNA microarrays are solid support grids of fixed short ssDNA fragments (oligonucleotides) that act as probes for tested genomic DNA targets. Highly complementary DNA hybridizes with the probe even under stringent conditions, while mismatched pairs are less stable. Probe-target hybridization is usually detected and quantified by detection of fluorescent, chemiluminescent or colorimetric signal. The intensity of the signal from each spot on the grid is determined under different hybridization conditions, and the relative abundance of each nucleic acid sequences in the target is identified, thus allowing the identification of subpopulations of sequences, like somatic mutations-baring fragments with low error frequency [151, 152]. This method allows the detection of low abundance point mutations in complex samples [76].

Multiplex ligation‐dependent probe amplification (MLPA)

In Multiplex Ligation-dependent Probe Amplification (MLPA) probe pairs hybridize specifically head-to-tail to target sequences in liquid media. Hybridized probes are subsequently ligated and amplified by PCR using fluorescent primers complementary to tag-sequences inserted in the probes (Figure 7A). PCR products are separated by capillary electrophoresis on an automated fragment analyzer, where peak heights indicate the amount of amplified product of each separate probe pair (Figure 7B). The absence or the poor amplification of specific probes means deletions, while unusually high amplification suggest gene duplications [153-157]. MLPA is a high throughput and cost-effective method that has been used in multiple studies of which only a few are referenced here [158-162].

Somatic Mutations  figure 7
Figure 7. Multiplex Ligation dependent Probe Amplification A) Schematic diagram; B) Typical capillary electrophoresis automated fragment analyzer result (modified from [3] ).
DNA and RNA in-situ hybridization (DISH and RISH)

In situ hybridization (ISH) can be used to determine the exact order of specific DNA and RNA fragments within a larger molecule. The technique is based on the ability of the double-stranded DNAs to denature to single-stranded upon heating and to pair with its complementary DNA or RNA sequence under non-denaturing conditions. When a labeled fragment of DNA (a DNA probe) is denatured and added to denatured DNA or RNA samples, some of the labeled DNA will hybridize to its complementary sequence. These duplexes are detected based on the detection of the probe, commonly by detection of its fluorescent signal. Two probes specific to sequences distant in the genome, which anneal at the same site in a chromosome, indicate a possible DNA junction. Based on this technique, exon-exon junctions were detected in short mRNA and gene variants of the APP gene associated with Alzheimer’s disease [45]. Such recombination events are somatic, and occur only in diseased neurons, but not in normal brains [45]. DNA sequencing of the probe-detected regions can confirm the junction.

Somatic Mutation Detection by Sequencing
Bulk sequencing-based methods

All somatic mutations can be identified by determining the complete nucleotide sequence of the genome. Several methods have been developed, starting from the first generation (classical) Sanger sequencing, to the more sophisticated next-generation approaches.

Direct sequencing

Direct sequencing is based on Sanger’s chain termination reaction technique [163]. It is a simple, accurate, and practical method for identifying mutations in genes associated with disease when the number of samples available is small. For example, multiple mutations were identified in the TCOF1 gene associated with Collins Treacher syndrome [164], mutations in the FGFR2 gene were detected in patients with Apert syndrome [165], while mutations in the PI3K [166] and KRAS [167] genes were identified in gallbladder and colon cancers using direct sequencing. More recently, direct sequencing was used in combination with locked-DNA PCR (LNA-PCR) to detect low-frequency mutations. Albitar et al [168] identified 1 mutant in a background of 1,000 wild type alleles by this method.

In spite of its simplicity, the method is not appropriate for the detection of unknown mutations when a large number of genes are candidate genes or when there is no candidate gene at all. It is also not considered the method of choice for detecting small deletions, but it is practical for the detection of mutations in genes involved in genetic disorders.

Next-generation (high-throughput) sequencing (NGS)

In contrast to the direct sequencing method devised by Sanger, next-generation sequencing involves the repeated sequencing of DNA segments randomly generated by genome fragmentation. The resulting sequence reads, of which many partly overlap, are aligned to a reference sequence (if available) and the full genome sequence is determined based on consensus [169, 170]. A few platforms for NGS have been developed and are used for both DNA and RNA sequencing. These include Illumina (Solexa) sequencing, Roche 454 sequencing, and Life Technologies Ion torrent: Proton / PGM sequencing or SOLiD sequencing, and were reviewed by Gundry and Vijk, 2012 [171, 172]. Alternatively, the full exosome [173, 174] or just specific targets are being sequence analyzed, especially for identification of mutations in monogenetic disorders [175]. The Agilent Sure Select Target enrichment system has been recently used for the detection of APP short gene variant in brains with Alzheimer’s disease [45].

All platforms allow the fast sequencing of entire genomes but are error-prone [36]. Because sequencing errors might not be distinguished from true mutations in the case of low-abundance somatic mutations, approaches to increase the sensitivity of the sequencing instruments were developed. Among them are Duplex Sequencing [176], Safe-Sequencing System ("Safe-SeqS") [36] and circle sequencing [4].

In duplex sequencing, the two strands of a DNA duplex are sequenced independently. As the two strands are complementary, true mutations are found in the same position in both strands [176]. Safe-SeqS is a barcoding method that involves the individual labeling of each DNA fragment, followed by amplification and the sequencing of the amplified product. Since multiple daughter molecules with identical sequence tags are generated, a preexistent mutation will be present in every daughter molecule containing the same tag in contrast to mutations introduced by sequencing or amplification which could be present in a much smaller subset of molecules [36]. In circular sequencing, DNA is denatured to single-stranded and circularized before performing rolling circle replication. Tandem-linked copies of the template circle are sequenced using any high-throughput sequencing technology, and the read sequence is computationally divided into individual copies of the original circle. If the first circle contains a mutation, the mutation will appear in multiple copies [4]. Figure 8 shows a comparative illustration of the above-described methods.

Somatic Mutations  figure 8
Figure 8. Overview of barcoding, circle and duplex sequencing (modified from [4].)

The several approaches available for analyzing somatic mutations apply mostly to point mutations and small insertions or deletions. Large deletions, insertions, inversions, or translocations cannot be detected by bulk DNA sequencing. An alternative sequencing method, ultra-low coverage sequencing, can be used to identify large somatic mutations by using a single sequencing read that spread across multiple reads as unique events [177].

Single-molecule real-time sequencing (SMTR sequencing)

In this technique for DNA sequencing, a single DNA polymerase or reverse-transcriptase enzyme and a single molecule of template DNA or RNA are localized at the bottom of an optical unit called zero-mode waveguide (ZMW). The ZMW contains a detector able to detect base-specific fluorescent signals from each nucleotide incorporated. The four bases are pre-labeled with specific fluorescent dyes, and upon their incorporation into the nascent DNA the tags are released. They diffuse out of the ZMW’s area where fluorescence is no longer observable. Thus, DNA synthesis can be observed in real-time. The technique developed by PacBio ( Smrt-seq was recently used by Lee et al to detect somatic variants of Alzheimer’s disease-associated gene APP [45]. In this study, both amplified DNA and cDNA fragments, as well as APP specific RNA were used as templates. Vasan N et al used PacBio SMRT-seq to resolve whether two PIK3CA mutations were on the same allele (cis configuration) or different alleles (trans configuration) [183].

Single-cell sequencing

Sequencing the genome of individual cells can reveal somatic mutations and facilitates the analysis of clonal evolution. To perform any single-cell sequencing assay, individual cells first have to be isolated from the system of interest. The most commonly used method is FACS. However, serial dilutions, microfluidic devices [184], optical tweezers [185, 186], and manual micromanipulation [187, 188] can also be used. All these methods have been reviewed elsewhere [189-193]. Methods for isolating single cells from rare cellular populations have also been developed and were reviewed recently by Navin NE (Figure 9) [5].

Somatic Mutations  figure 9
Figure 9. Summary of single-cell sorting methods reviewed by Navin, NE [5]. A) Methods for isolating cells from abundant-cell populations. B) Methods from isolating cells from rare-cell populations.

Rare cells have been enriched from circulating tumors by CellSearch (Johnson and Johnson) [194], isolated by DEPArray (Silicon Biosciences) [195], and their whole genomes were amplified and analyzed by NGS (Next-Generation Sequencing) [196, 197] or by Sanger sequencing [198, 199] to identify somatic mutations in the genome. Other studies have used whole genome amplification after single cell isolation by laser microdissection [178, 200-202] and different sequencing strategies [200, 201, 203]. Some skipped the whole genome amplification step altogether [204]. A different technology, called MagSweeper (Illumina) that uses a rotating magnet with bound EpCAM antibodies to isolate circulating tumor cells [205] was used in combination with DNA sequencing [206] to detect somatic mutations in tumor cells in breast cancers.

An alternative to genome sequencing for somatic mutation detection from single cells is the sequencing of the cell’s transcriptome through RNA-seq, as it can provide information at single nucleotide resolution. Mutations can be identified directly by RNA-seq [207, 208] or validated after their detection by DNA-seq or other methods [209]. RNA-seq is often the sequencing method of choice due to budget limitations, sample quantities or study goals. However, analyzing RNA-seq data can be challenging because it can have a high false positive rate for single nucleotide variations [210, 211]. This is a consequence of the biology of RNA metabolism (e.g., RNA editing, RNA splicing), as well as of the molecular techniques (e.g., errors introduced during reverse transcription and PCR) and sequence analysis methods (e.g., strand bias [211, 212], alignment complexity [213] ).

Most RNA-seq data specific variant detection tools were developed for single nucleotide variation detection rather than for somatic mutations [213, 214], but more recent devices are designed and used to identify somatic mutations [208, 215-217].

Yizhak K et al adapted the MuTect platform [218] to identify somatic mutations from large RNA-seq databases Cancer Genome Atlas and Genotype-Tissue Expression (GTEx) project, validated their approach through Fluidigm microfluidic PCR and the Illumina MiSeq sequencing system, and identified skin, lung, and esophagus as most frequent tissues for somatic mutations [219].

Strategies for Detecting Somatic Mutations in Diseased Tissues

Somatic mutations can be identified from parallel sequencing data by directly comparing the DNA sequence from tumor samples with their normal counterparts. The paired tumor/normal approach permits the identification and elimination of genomic variants due to their presence in all the cells of an individual. Somatic mutations are identified based on their existence only in a subset of cells. For example, T Powles et al identified somatic variants based on the tumour tissue and matched normal sequencing data and used those variants as the markers to detect the presence of circulating tumour DNA [83]. Commonly mutated sites have been identified in a variety of cancers by using this method [220]. One inconvenience of this approach is the need to sequence twice as many samples as the number of test samples, leading to higher than needed costs and analysis duration. Also, for each test sample, a matching standard tissue sample is required, and sometimes this might not be available.

Recently, an alternative approach, in which only test samples are used, has been developed [221], but data suggest that matched tumor-normal sequencing is needed for precise identification of somatic mutations [222]. A slightly improved normal-tissue free analysis technique was developed by Jamie et al [223]. This method uses a specialized Genome Analysis Tool Kit (GATK) pipeline [224] to improve the detection of somatic mutations.

References
  1. Quan P, Sauzade M, Brouzes E. dPCR: A Technology Review. Sensors (Basel). 2018;18: pubmed publisher
  2. Amicarelli G, Shehi E, Makrigiorgos G, Adlerstein D. FLAG assay as a novel method for real-time signal generation during PCR: application to detection and genotyping of KRAS codon 12 mutations. Nucleic Acids Res. 2007;35:e131 pubmed
  3. Harteveld C, Higgs D. Alpha-thalassaemia. Orphanet J Rare Dis. 2010;5:13 pubmed publisher
  4. Lou D, Hussmann J, McBee R, Acevedo A, Andino R, Press W, et al. High-throughput DNA sequencing errors are reduced by orders of magnitude using circle sequencing. Proc Natl Acad Sci U S A. 2013;110:19872-7 pubmed publisher
  5. Navin N. Cancer genomics: one cell at a time. Genome Biol. 2014;15:452 pubmed publisher
  6. Baez Ortega A, Gori K, Strakova A, Allen J, Allum K, Bansse Issa L, et al. Somatic evolution and global expansion of an ancient transmissible cancer lineage. Science. 2019;365: pubmed publisher
  7. Martincorena I, Roshan A, Gerstung M, Ellis P, Van Loo P, McLaren S, et al. Tumor evolution. High burden and pervasive positive selection of somatic mutations in normal human skin. Science. 2015;348:880-6 pubmed publisher
  8. Xia L, Li Z, Zhou B, Tian G, Zeng L, Dai H, et al. Statistical analysis of mutant allele frequency level of circulating cell-free DNA and blood cells in healthy individuals. Sci Rep. 2017;7:7526 pubmed publisher
  9. Hall J. Review and hypotheses: somatic mosaicism: observations related to clinical genetics. Am J Hum Genet. 1988;43:355-63 pubmed
  10. Gajecka M. Unrevealed mosaicism in the next-generation sequencing era. Mol Genet Genomics. 2016;291:513-30 pubmed publisher
  11. Cagan A, Baez Ortega A, Brzozowska N, Abascal F, Coorens T, Sanders M, et al. Somatic mutation rates scale with lifespan across mammals. Nature. 2022;604:517-524 pubmed publisher
  12. Nanki K, Fujii M, Shimokawa M, Matano M, Nishikori S, Date S, et al. Somatic inflammatory gene mutations in human ulcerative colitis epithelium. Nature. 2019;: pubmed publisher
  13. Zhang L, Dong X, Lee M, Maslov A, Wang T, Vijg J. Single-cell whole-genome sequencing reveals the functional landscape of somatic mutations in B lymphocytes across the human lifespan. Proc Natl Acad Sci U S A. 2019;116:9014-9019 pubmed publisher
  14. Yoshida K, Gowers K, Lee Six H, Chandrasekharan D, Coorens T, Maughan E, et al. Tobacco smoking and somatic mutations in human bronchial epithelium. Nature. 2020;578:266-272 pubmed publisher
  15. De S. Somatic mosaicism in healthy human tissues. Trends Genet. 2011;27:217-23 pubmed publisher
  16. Frank S. Somatic mosaicism and disease. Curr Biol. 2014;24:R577-R581 pubmed publisher
  17. Risques R, Kennedy S. Aging and the rise of somatic cancer-associated mutations in normal tissues. PLoS Genet. 2018;14:e1007108 pubmed publisher
  18. Martincorena I, Campbell P. Somatic mutation in cancer and normal cells. Science. 2015;349:1483-9 pubmed publisher
  19. Gold B. Somatic mutations in cancer: Stochastic versus predictable. Mutat Res. 2017;814:37-46 pubmed publisher
  20. Martincorena I, Raine K, Gerstung M, Dawson K, Haase K, Van Loo P, et al. Universal Patterns of Selection in Cancer and Somatic Tissues. Cell. 2017;171:1029-1041.e21 pubmed publisher
  21. Kent D, Green A. Order Matters: The Order of Somatic Mutations Influences Cancer Evolution. Cold Spring Harb Perspect Med. 2017;7: pubmed publisher
  22. Lee J. Somatic mutations in disorders with disrupted brain connectivity. Exp Mol Med. 2016;48:e239 pubmed publisher
  23. Mass E, Jacome Galarza C, Blank T, Lazarov T, Durham B, Ozkaya N, et al. A somatic mutation in erythro-myeloid progenitors causes neurodegenerative disease. Nature. 2017;549:389-393 pubmed publisher
  24. Clark V, Harmanci A, Bai H, Youngblood M, Lee T, Baranoski J, et al. Recurrent somatic mutations in POLR2A define a distinct subset of meningiomas. Nat Genet. 2016;48:1253-9 pubmed publisher
  25. McConnell M, Moran J, Abyzov A, Akbarian S, Bae T, Cortes Ciriano I, et al. Intersection of diverse neuronal genomes and neuropsychiatric disease: The Brain Somatic Mosaicism Network. Science. 2017;356: pubmed publisher
  26. Erickson R. Recent advances in the study of somatic mosaicism and diseases other than cancer. Curr Opin Genet Dev. 2014;26:73-8 pubmed publisher
  27. Stephens P, McBride D, Lin M, Varela I, Pleasance E, Simpson J, et al. Complex landscapes of somatic rearrangement in human breast cancer genomes. Nature. 2009;462:1005-10 pubmed publisher
  28. Alexandrov L, Nik Zainal S, Wedge D, Aparicio S, Behjati S, Biankin A, et al. Signatures of mutational processes in human cancer. Nature. 2013;500:415-21 pubmed publisher
  29. Lawrence M, Stojanov P, Polak P, Kryukov G, Cibulskis K, Sivachenko A, et al. Mutational heterogeneity in cancer and the search for new cancer-associated genes. Nature. 2013;499:214-218 pubmed publisher
  30. Vogelstein B, Papadopoulos N, Velculescu V, Zhou S, Diaz L, Kinzler K. Cancer genome landscapes. Science. 2013;339:1546-58 pubmed publisher
  31. Tate J, Bamford S, Jubb H, Sondka Z, Beare D, Bindal N, et al. COSMIC: the Catalogue Of Somatic Mutations In Cancer. Nucleic Acids Res. 2019;47:D941-D947 pubmed publisher
  32. Verschueren E, Husain B, Yuen K, Sun Y, Paduchuri S, Senbabaoglu Y, et al. The Immunoglobulin Superfamily Receptome Defines Cancer-Relevant Networks Associated with Clinical Outcome. Cell. 2020;182:329-344.e19 pubmed publisher
  33. Yu K, Lin C, Hatcher A, Lozzi B, Kong K, Huang Hobbs E, et al. PIK3CA variants selectively initiate brain hyperactivity during gliomagenesis. Nature. 2020;578:166-171 pubmed publisher
  34. Wan L, Chong S, Xuan F, Liang A, Cui X, Gates L, et al. Impaired cell fate through gain-of-function mutations in a chromatin reader. Nature. 2020;577:121-126 pubmed publisher
  35. Zacharakis N, Chinnasamy H, Black M, Xu H, Lu Y, Zheng Z, et al. Immune recognition of somatic mutations leading to complete durable regression in metastatic breast cancer. Nat Med. 2018;24:724-730 pubmed publisher
  36. Kinde I, Wu J, Papadopoulos N, Kinzler K, Vogelstein B. Detection and quantification of rare mutations with massively parallel sequencing. Proc Natl Acad Sci U S A. 2011;108:9530-5 pubmed publisher
  37. Balderston S, Taulbee J, Celaya E, Fung K, Jiao A, Smith K, et al. Discrimination of single-point mutations in unamplified genomic DNA via Cas9 immobilized on a graphene field-effect transistor. Nat Biomed Eng. 2021;: pubmed publisher
  38. Newton C, Graham A, Heptinstall L, Powell S, Summers C, Kalsheker N, et al. Analysis of any point mutation in DNA. The amplification refractory mutation system (ARMS). Nucleic Acids Res. 1989;17:2503-16 pubmed
  39. Milbury C, Li J, Makrigiorgos G. PCR-based methods for the enrichment of minority alleles and mutations. Clin Chem. 2009;55:632-40 pubmed publisher
  40. Alyethodi R, Singh U, Kumar S, Alex R, Deb R, Sengar G, et al. T-ARMS PCR genotyping of SNP rs445709131 using thermostable strand displacement polymerase. BMC Res Notes. 2018;11:132 pubmed publisher
  41. Yang L, Ijaz I, Cheng J, Wei C, Tan X, Khan M, et al. Evaluation of amplification refractory mutation system (ARMS) technique for quick and accurate prenatal gene diagnosis of CHM variant in choroideremia. Appl Clin Genet. 2018;11:1-8 pubmed publisher
  42. Ryan S, Ryan F, O Dwyer V, Neylan D. A real-time ARMS PCR/high-resolution melt curve assay for the detection of the three primary mitochondrial mutations in Leber's hereditary optic neuropathy. Mol Vis. 2016;22:1169-1175 pubmed
  43. Donà V, Smid J, Kasraian S, Egli Gany D, Dost F, Imeri F, et al. Mismatch Amplification Mutation Assay-Based Real-Time PCR for Rapid Detection of Neisseria gonorrhoeae and Antimicrobial Resistance Determinants in Clinical Specimens. J Clin Microbiol. 2018;56: pubmed publisher
  44. Hu Y, Duan Q, Chen Y, Yao L, Chen Z, Li K, et al. A novel multiplex rt-PCR assay for the detection of four chromosomal translocations of leukemia. Genet Test Mol Biomarkers. 2014;18:810-9 pubmed publisher
  45. Lee M, Siddoway B, Kaeser G, Segota I, Rivera R, Romanow W, et al. Somatic APP gene recombination in Alzheimer's disease and normal neurons. Nature. 2018;563:639-645 pubmed publisher
  46. Chen S, Zhao J, Cui L, Liu Y. Urinary circulating DNA detection for dynamic tracking of EGFR mutations for NSCLC patients treated with EGFR-TKIs. Clin Transl Oncol. 2017;19:332-340 pubmed publisher
  47. Takeshita T, Yamamoto Y, Yamamoto Ibusuki M, Inao T, Sueta A, Fujiwara S, et al. Clinical significance of monitoring ESR1 mutations in circulating cell-free DNA in estrogen receptor positive breast cancer patients. Oncotarget. 2016;7:32504-18 pubmed publisher
  48. Chang Y, Er T, Lu H, Yeh K, Chang J. Detection of KRAS codon 12 and 13 mutations by mutant-enriched PCR assay. Clin Chim Acta. 2014;436:169-75 pubmed publisher
  49. Däbritz J, Hänfler J, Preston R, Stieler J, Oettle H. Detection of Ki-ras mutations in tissue and plasma samples of patients with pancreatic cancer using PNA-mediated PCR clamping and hybridisation probes. Br J Cancer. 2005;92:405-12 pubmed
  50. Zeng Q, Xie L, Zhou N, Liu M, Song X. Detection of PIK3CA Mutations in Plasma DNA of Colorectal Cancer Patients by an Ultra-Sensitive PNA-Mediated PCR. Mol Diagn Ther. 2017;21:443-451 pubmed publisher
  51. Kwon M, Lee S, Kang S, Choi Y. Frequency of KRAS, BRAF, and PIK3CA mutations in advanced colorectal cancers: Comparison of peptide nucleic acid-mediated PCR clamping and direct sequencing in formalin-fixed, paraffin-embedded tissue. Pathol Res Pract. 2011;207:762-8 pubmed publisher
  52. Oldenburg R, Liu M, Kolodney M. Selective amplification of rare mutations using locked nucleic acid oligonucleotides that competitively inhibit primer binding to wild-type DNA. J Invest Dermatol. 2008;128:398-402 pubmed
  53. Emelyanova M, Ghukasyan L, Abramov I, Ryabaya O, Stepanova E, Kudryavtseva A, et al. Detection of BRAF, NRAS, KIT, GNAQ, GNA11 and MAP2K1/2 mutations in Russian melanoma patients using LNA PCR clamp and biochip analysis. Oncotarget. 2017;8:52304-52320 pubmed publisher
  54. Abdelhamid E, Besbes S, Renneville A, Nibourel O, Helevaut N, Preudhomme C, et al. Minimal Residual Disease assessment of IDH1/2 mutations in Acute Myeloid Leukemia by LNA-RQ-PCR. Tunis Med. 2016;94:190-7 pubmed
  55. Li J, Wang L, Mamon H, Kulke M, Berbeco R, Makrigiorgos G. Replacing PCR with COLD-PCR enriches variant DNA sequences and redefines the sensitivity of genetic testing. Nat Med. 2008;14:579-84 pubmed publisher
  56. Milbury C, Correll M, Quackenbush J, Rubio R, Makrigiorgos G. COLD-PCR enrichment of rare cancer mutations prior to targeted amplicon resequencing. Clin Chem. 2012;58:580-9 pubmed publisher
  57. Paganini I, Mancini I, Baroncelli M, Arena G, Gensini F, Papi L, et al. Application of COLD-PCR for improved detection of NF2 mosaic mutations. J Mol Diagn. 2014;16:393-9 pubmed publisher
  58. Kakavas V, Konstantinos K, Plageras P, Panagiotis P, Vlachos T, Antonios V, et al. PCR-SSCP: a method for the molecular analysis of genetic diseases. Mol Biotechnol. 2008;38:155-63 pubmed publisher
  59. Nataraj A, Olivos Glander I, Kusukawa N, Highsmith W. Single-strand conformation polymorphism and heteroduplex analysis for gel-based mutation detection. Electrophoresis. 1999;20:1177-85 pubmed
  60. Rabalski L, Smietanka K, Minta Z, Szewczyk B. Detection of Newcastle disease virus minor genetic variants by modified single-stranded conformational polymorphism analysis. Biomed Res Int. 2014;2014:632347 pubmed publisher
  61. Fodde R, Losekoot M. Mutation detection by denaturing gradient gel electrophoresis (DGGE). Hum Mutat. 1994;3:83-94 pubmed
  62. Li J, Xin J, Zhang L, Jiang L, Cao H, Li L. Rapid detection of rpoB mutations in rifampin resistant M. tuberculosis from sputum samples by denaturing gradient gel electrophoresis. Int J Med Sci. 2012;9:148-56 pubmed publisher
  63. Liu W, Li B, Chu H, Zhang Z, Luo L, Ma W, et al. Rapid detection of mutations in erm(41) and rrl associated with clarithromycin resistance in Mycobacterium abscessus complex by denaturing gradient gel electrophoresis. J Microbiol Methods. 2017;143:87-93 pubmed publisher
  64. Ekstrøm P, Børresen Dale A, Qvist H, Giercksky K, Thilly W. Detection of low-frequency mutations in exon 8 of the TP53 gene by constant denaturant capillary electrophoresis (CDCE). Biotechniques. 1999;27:128-34 pubmed
  65. Khrapko K, Coller H, Li Sucholeiki X, André P, Thilly W. High resolution analysis of point mutations by constant denaturant capillary electrophoresis (CDCE). Methods Mol Biol. 2001;163:57-72 pubmed
  66. Fält S, Kumar R, Wennborg A, Tomita Mitchell A, Thilly W, Lambert B. Identification of in vivo mutations in exon 5 of the human HPRT gene in a set of pooled T-cell mutants by constant denaturant capillary electrophoresis (CDCE). Mutat Res. 2000;452:57-66 pubmed
  67. Liu W, Smith D, Rechtzigel K, Thibodeau S, James C. Denaturing high performance liquid chromatography (DHPLC) used in the detection of germline and somatic mutations. Nucleic Acids Res. 1998;26:1396-400 pubmed
  68. Klein B, Weirich G, Brauch H. DHPLC-based germline mutation screening in the analysis of the VHL tumor suppressor gene: usefulness and limitations. Hum Genet. 2001;108:376-84 pubmed
  69. Gentilini F, Mantovani V, Turba M. The use of COLD-PCR, DHPLC and GeneScanning for the highly sensitive detection of c-KIT somatic mutations in canine mast cell tumours. Vet Comp Oncol. 2015;13:218-28 pubmed publisher
  70. Kan Z, Jaiswal B, Stinson J, Janakiraman V, Bhatt D, Stern H, et al. Diverse somatic mutation patterns and pathway alterations in human cancers. Nature. 2010;466:869-73 pubmed publisher
  71. Haliassos A, Chomel J, Grandjouan S, Kruh J, Kaplan J, Kitzis A. Detection of minority point mutations by modified PCR technique: a new approach for a sensitive diagnosis of tumor-progression markers. Nucleic Acids Res. 1989;17:8093-9 pubmed
  72. Gonzalez Bosquet J, Calcei J, Wei J, Garcia Closas M, Sherman M, Hewitt S, et al. Detection of somatic mutations by high-resolution DNA melting (HRM) analysis in multiple cancers. PLoS ONE. 2011;6:e14522 pubmed publisher
  73. Ney J, Froehner S, Roesler A, Buettner R, Merkelbach Bruse S. High-resolution melting analysis as a sensitive prescreening diagnostic tool to detect KRAS , BRAF , PIK3CA , and AKT1 mutations in formalin-fixed, paraffin-embedded tissues. Arch Pathol Lab Med. 2012;136:983-92 pubmed publisher
  74. Sutton B, Birse R, Maggert K, Ray T, Hobbs J, Ezenekwe A, et al. Assessment of common somatic mutations of EGFR, KRAS, BRAF, NRAS in pulmonary non-small cell carcinoma using iPLEX® HS, a new highly sensitive assay for the MassARRAY® System. PLoS ONE. 2017;12:e0183715 pubmed publisher
  75. Spaans V, Trietsch M, Crobach S, Stelloo E, Kremer D, Osse E, et al. Designing a high-throughput somatic mutation profiling panel specifically for gynaecological cancers. PLoS ONE. 2014;9:e93451 pubmed publisher
  76. Pingel J, Buhot A, Calemczuk R, Livache T. Temperature scans/cycles for the detection of low abundant DNA point mutations on microarrays. Biosens Bioelectron. 2012;31:554-7 pubmed publisher
  77. Cusumano A, Busin M, Spitznas M, Koch F. Epikeratophakia for the correction of myopia: lenticule design and related histopathological findings. Refract Corneal Surg. 1990;6:120-4 pubmed
  78. Veldhuisen B, van der Schoot C, de Haas M. Multiplex ligation-dependent probe amplification (MLPA) assay for blood group genotyping, copy number quantification, and analysis of RH variants. Immunohematology. 2015;31:58-61 pubmed
  79. Deepha S, Vengalil S, Preethish Kumar V, Polavarapu K, Nalini A, Gayathri N, et al. MLPA identification of dystrophin mutations and in silico evaluation of the predicted protein in dystrophinopathy cases from India. BMC Med Genet. 2017;18:67 pubmed publisher
  80. Park H, Park S, Im K, Kim S, Kim J, Hwang S, et al. Telomere length and somatic mutations in correlation with response to immunosuppressive treatment in aplastic anaemia. Br J Haematol. 2017;178:603-615 pubmed publisher
  81. Lambo S, Gröbner S, Rausch T, Waszak S, Schmidt C, Gorthi A, et al. The molecular landscape of ETMR at diagnosis and relapse. Nature. 2019;576:274-280 pubmed publisher
  82. Petri K, Zhang W, Ma J, Schmidts A, Lee H, Horng J, et al. CRISPR prime editing with ribonucleoprotein complexes in zebrafish and primary human cells. Nat Biotechnol. 2021;: pubmed publisher
  83. Powles T, Assaf Z, Davarpanah N, Banchereau R, Szabados B, Yuen K, et al. ctDNA guiding adjuvant immunotherapy in urothelial carcinoma. Nature. 2021;595:432-437 pubmed publisher
  84. Nam A, Kim K, Chaligne R, Izzo F, Ang C, Taylor J, et al. Somatic mutations and cell identity linked by Genotyping of Transcriptomes. Nature. 2019;571:355-360 pubmed publisher
  85. Lanz T, Brewer R, Ho P, Moon J, Jude K, Fernandez D, et al. Clonally Expanded B Cells in Multiple Sclerosis Bind EBV EBNA1 and GlialCAM. Nature. 2022;: pubmed publisher
  86. Bottema C, Sommer S. PCR amplification of specific alleles: rapid detection of known mutations and polymorphisms. Mutat Res. 1993;288:93-102 pubmed
  87. Sommer S, Groszbach A, Bottema C. PCR amplification of specific alleles (PASA) is a general method for rapidly detecting known single-base changes. Biotechniques. 1992;12:82-7 pubmed
  88. Zschocke J, Graham C. A fluorescent multiplex ARMS method for rapid mutation analysis. Mol Cell Probes. 1995;9:447-51 pubmed
  89. Maher C, Crowley D, Cullen C, Wall C, Royston D, Fanning S. Double fluorescent-amplification refractory mutation detection (dF-ARMS) of the factor V Leiden and prothrombin mutations. Thromb Haemost. 1999;81:76-80 pubmed
  90. Ririe K, Rasmussen R, Wittwer C. Product differentiation by analysis of DNA melting curves during the polymerase chain reaction. Anal Biochem. 1997;245:154-60 pubmed
  91. Niederstätter H, Parson W. Fluorescent duplex allele-specific PCR and amplicon melting for rapid homogeneous mtDNA haplogroup H screening and sensitive mixture detection. PLoS ONE. 2009;4:e8374 pubmed publisher
  92. Wang C, Aleksic A, Xu M, Procyshyn R, Ross C, Vila Rodriguez F, et al. A Tetra-Primer Amplification Refractory System Technique for the Cost-Effective and Novel Genotyping of Eight Single-Nucleotide Polymorphisms of the Catechol-O-Methyltransferase Gene. Genet Test Mol Biomarkers. 2016;20:465-70 pubmed publisher
  93. Randhawa R, Duseja A, Changotra H. A novel Tetra-primer ARMS-PCR based assay for genotyping SNP rs12303764(G/T) of human Unc-51 like kinase 1 gene. Mol Biol Rep. 2017;44:1-4 pubmed publisher
  94. Suhda S, Paramita D, Fachiroh J. Tetra Primer ARMS PCR Optimization to Detect Single Nucleotide Polymorphisms of the CYP2E1 Gene. Asian Pac J Cancer Prev. 2016;17:3065-9 pubmed
  95. Gibbs R, Nguyen P, Caskey C. Detection of single DNA base differences by competitive oligonucleotide priming. Nucleic Acids Res. 1989;17:2437-48 pubmed
  96. Kahn S, Jiang W, Culbertson T, Weinstein I, Williams G, Tomita N, et al. Rapid and sensitive nonradioactive detection of mutant K-ras genes via 'enriched' PCR amplification. Oncogene. 1991;6:1079-83 pubmed
  97. Ronai Z, Minamoto T. Quantitative enriched PCR (QEPCR), a highly sensitive method for detection of K-ras oncogene mutation. Hum Mutat. 1997;10:322-5 pubmed
  98. Cha R, Zarbl H, Keohavong P, Thilly W. Mismatch amplification mutation assay (MAMA): application to the c-H-ras gene. PCR Methods Appl. 1992;2:14-20 pubmed
  99. Takeda S, Ichii S, Nakamura Y. Detection of K-ras mutation in sputum by mutant-allele-specific amplification (MASA). Hum Mutat. 1993;2:112-7 pubmed
  100. Myers M, McKinzie P, Wang Y, Meng F, Parsons B. ACB-PCR quantification of somatic oncomutation. Methods Mol Biol. 2014;1105:345-63 pubmed publisher
  101. Ntziora F, Paraskevis D, Haida C, Manesis E, Papatheodoridis G, Manolakopoulos S, et al. Ultrasensitive amplification refractory mutation system real-time PCR (ARMS RT-PCR) assay for detection of minority hepatitis B virus-resistant strains in the era of personalized medicine. J Clin Microbiol. 2013;51:2893-900 pubmed publisher
  102. Miranzadeh Mahabadi H, Miranzadeh Mahabadi H, Nikpour P, Emadi Baygi M, Kelishadi R. Comparison of TaqMan Real-Time and Tetra-Primer ARMS PCR Techniques for Genotyping of Rs 8066560 Variant in Children and Adolescents with Metabolic Syndrome. Adv Clin Exp Med. 2015;24:951-5 pubmed publisher
  103. Sabui S, Dutta S, Debnath A, Ghosh A, Hamabata T, Rajendran K, et al. Real-time PCR-based mismatch amplification mutation assay for specific detection of CS6-expressing allelic variants of enterotoxigenic Escherichia coli and its application in assessing diarrheal cases and asymptomatic controls. J Clin Microbiol. 2012;50:1308-12 pubmed publisher
  104. Wolff J, Gemmell N. Combining allele-specific fluorescent probes and restriction assay in real-time PCR to achieve SNP scoring beyond allele ratios of 1:1000. Biotechniques. 2008;44:193-4, 196, 199 pubmed
  105. Bustin S, Benes V, Garson J, Hellemans J, Huggett J, Kubista M, et al. The MIQE guidelines: minimum information for publication of quantitative real-time PCR experiments. Clin Chem. 2009;55:611-22 pubmed publisher
  106. Alonso C, Gallego M, Rossi J, Medina A, Rubio P, Bernasconi A, et al. RT-PCR diagnosis of recurrent rearrangements in pediatric acute lymphoblastic leukemia in Argentina. Leuk Res. 2012;36:704-8 pubmed publisher
  107. Couto J, Huang A, Konczyk D, Goss J, Fishman S, Mulliken J, et al. Somatic MAP2K1 Mutations Are Associated with Extracranial Arteriovenous Malformation. Am J Hum Genet. 2017;100:546-554 pubmed publisher
  108. Luks V, Kamitaki N, Vivero M, Uller W, Rab R, Bovée J, et al. Lymphatic and other vascular malformative/overgrowth disorders are caused by somatic mutations in PIK3CA. J Pediatr. 2015;166:1048-54.e1-5 pubmed publisher
  109. Nielsen P, Egholm M, Berg R, Buchardt O. Sequence-selective recognition of DNA by strand displacement with a thymine-substituted polyamide. Science. 1991;254:1497-500 pubmed
  110. Hong C, Yang C, Zhuang Z. Application of Peptide Nucleic Acid-based Assays Toward Detection of Somatic Mosaicism. Mol Ther Nucleic Acids. 2016;5:e314 pubmed publisher
  111. Dominguez P, Kolodney M. Wild-type blocking polymerase chain reaction for detection of single nucleotide minority mutations from clinical specimens. Oncogene. 2005;24:6830-4 pubmed
  112. Zuo Z, Jabbar K. COLD-PCR: Applications and Advantages. Methods Mol Biol. 2016;1392:17-25 pubmed publisher
  113. Mauger F, How Kit A, Tost J. COLD-PCR Technologies in the Area of Personalized Medicine: Methodology and Applications. Mol Diagn Ther. 2017;21:269-283 pubmed publisher
  114. Ghalamkari S, Khosravian F, Mianesaz H, Kazemi M, Behjati M, Hakimian S, et al. A Comparison Between Full-COLD PCR/HRM and PCR Sequencing for Detection of Mutations in Exon 9 of PIK3CA in Breast Cancer Patients. Appl Biochem Biotechnol. 2019;187:975-983 pubmed publisher
  115. Wong D, Fung J, Lai C, Yuen M. COLD-PCR for early detection of hepatitis B virus antiviral drug resistance mutations. Hong Kong Med J. 2015;21 Suppl 7:S8-10 pubmed
  116. Song C, Milbury C, Li J, Liu P, Zhao M, Makrigiorgos G. Rapid and sensitive detection of KRAS mutation after fast-COLD-PCR enrichment and high-resolution melting analysis. Diagn Mol Pathol. 2011;20:81-9 pubmed publisher
  117. Carotenuto P, Roma C, Cozzolino S, Fenizia F, Rachiglio A, Tatangelo F, et al. Detection of KRAS mutations in colorectal cancer with Fast COLD-PCR. Int J Oncol. 2012;40:378-84 pubmed publisher
  118. Orita M, Iwahana H, Kanazawa H, Hayashi K, Sekiya T. Detection of polymorphisms of human DNA by gel electrophoresis as single-strand conformation polymorphisms. Proc Natl Acad Sci U S A. 1989;86:2766-70 pubmed
  119. Delaunay A, Dallot S, Filloux D, Dupuy V, Roumagnac P, Jacquot E. SNaPshot and CE-SSCP: Two Simple and Cost-Effective Methods to Reveal Genetic Variability Within a Virus Species. Methods Mol Biol. 2015;1302:187-206 pubmed publisher
  120. Woo N, Kim S, Kang S. Voltage-programming-based capillary gel electrophoresis for the fast detection of angiotensin-converting enzyme insertion/deletion polymorphism with high sensitivity. J Sep Sci. 2016;39:3230-8 pubmed publisher
  121. Sørlie T, Johnsen H, Vu P, Lind G, Lothe R, Børresen Dale A. Mutation screening of the TP53 gene by temporal temperature gel electrophoresis (TTGE). Methods Mol Biol. 2014;1105:315-24 pubmed publisher
  122. Xue M, Bonny O, Morgenthaler S, Bochud M, Mooser V, Thilly W, et al. Use of constant denaturant capillary electrophoresis of pooled blood samples to identify single-nucleotide polymorphisms in the genes (Scnn1a and Scnn1b) encoding the alpha and beta subunits of the epithelial sodium channel. Clin Chem. 2002;48:718-28 pubmed
  123. Khrapko K, Hanekamp J, Thilly W, Belenkii A, Foret F, Karger B. Constant denaturant capillary electrophoresis (CDCE): a high resolution approach to mutational analysis. Nucleic Acids Res. 1994;22:364-9 pubmed
  124. Kristensen A, Bjørheim J, Ekstrøm P. Detection of mutations in exon 8 of TP53 by temperature gradient 96-capillary array electrophoresis. Biotechniques. 2002;33:650-3 pubmed
  125. Li Sucholeiki X, Hu G, Perls T, Tomita Mitchell A, Thilly W. Scanning the beta-globin gene for mutations in large populations by denaturing capillary and gel electrophoresis. Electrophoresis. 2005;26:2531-8 pubmed
  126. Keller G, Hartmann A, Mueller J, Hofler H. Denaturing high pressure liquid chromatography (DHPLC) for the analysis of somatic p53 mutations. Lab Invest. 2001;81:1735-7 pubmed
  127. Luquin N, Yu B, Trent R, Pamphlett R. DHPLC can be used to detect low-level mutations in amyotrophic lateral sclerosis. Amyotroph Lateral Scler. 2010;11:76-82 pubmed publisher
  128. Chotirat S, Thongnoppakhun W, Wanachiwanawin W, Auewarakul C. Acquired somatic mutations of isocitrate dehydrogenases 1 and 2 (IDH1 and IDH2) in preleukemic disorders. Blood Cells Mol Dis. 2015;54:286-91 pubmed publisher
  129. Yu J, Yu S, Wang S, Bai H, Zhao J, An T, et al. Clinical outcomes of EGFR-TKI treatment and genetic heterogeneity in lung adenocarcinoma patients with EGFR mutations on exons 19 and 21. Chin J Cancer. 2016;35:30 pubmed publisher
  130. Upadhyaya M, Han S, Consoli C, Majounie E, Horan M, Thomas N, et al. Characterization of the somatic mutational spectrum of the neurofibromatosis type 1 (NF1) gene in neurofibromatosis patients with benign and malignant tumors. Hum Mutat. 2004;23:134-46 pubmed
  131. Wagner R, Debbie P, Radman M. Mutation detection using immobilized mismatch binding protein (MutS). Nucleic Acids Res. 1995;23:3944-8 pubmed
  132. Wagner R, Dean A. The use of immobilized mismatch binding protein in mutation/SNP detection. Methods Mol Biol. 2000;152:159-68 pubmed
  133. Bi L, Zhou Y, Zhang X, Deng J, Zhang Z, Xie B, et al. A MutS-based protein chip for detection of DNA mutations. Anal Chem. 2003;75:4113-9 pubmed
  134. Zhang X, Bi L. Protein chip for detection of DNA mutations. Methods Mol Biol. 2007;382:163-76 pubmed publisher
  135. Faham M, Cox D. A novel in vivo method to detect DNA sequence variation. Genome Res. 1995;5:474-82 pubmed
  136. Faham M, Zheng J, Moorhead M, Fakhrai Rad H, Namsaraev E, Wong K, et al. Multiplexed variation scanning for 1,000 amplicons in hundreds of patients using mismatch repair detection (MRD) on tag arrays. Proc Natl Acad Sci U S A. 2005;102:14717-22 pubmed
  137. Peters B, Kan Z, Sebisanovic D, Pujara K, Wang Z, Hong P, et al. Highly efficient somatic-mutation identification using Escherichia coli mismatch-repair detection. Nat Methods. 2007;4:713-5 pubmed
  138. Bentivegna S, Zheng J, Namsaraev E, Carlton V, Pavlicek A, Moorhead M, et al. Rapid identification of somatic mutations in colorectal and breast cancer tissues using mismatch repair detection (MRD). Hum Mutat. 2008;29:441-50 pubmed publisher
  139. Ward R, Hawkins N, O Grady R, Sheehan C, O CONNOR T, Impey H, et al. Restriction endonuclease-mediated selective polymerase chain reaction: a novel assay for the detection of K-ras mutations in clinical samples. Am J Pathol. 1998;153:373-9 pubmed
  140. Kaur M, Zhang Y, Liu W, Tetradis S, Price B, Makrigiorgos G. Ligation of a primer at a mutation: a method to detect low level mutations in DNA. Mutagenesis. 2002;17:365-74 pubmed
  141. Zhao J, Xie F, Zhong W, Wu W, Qu S, Gao S, et al. Restriction endonuclease-mediated real-time digestion-PCR for somatic mutation detection. Int J Cancer. 2013;132:2858-66 pubmed publisher
  142. Pourzand C, Cerutti P. Genotypic mutation analysis by RFLP/PCR. Mutat Res. 1993;288:113-21 pubmed
  143. Bujko M, Kober P, Matyja E, Nauman P, Dyttus Cebulok K, Czeremszyńska B, et al. Prognostic value of IDH1 mutations identified with PCR-RFLP assay in glioblastoma patients. Mol Diagn Ther. 2010;14:163-9 pubmed publisher
  144. Elsayed G, Nassar H, Zaher A, Elnoshokaty E, Moneer M. Prognostic value of IDH1 mutations identified with PCR-RFLP assay in acute myeloid leukemia patients. J Egypt Natl Canc Inst. 2014;26:43-9 pubmed publisher
  145. Trifa A, Cucuianu A, Popp R. Development of a reliable PCR-RFLP assay for investigation of the JAK2 rs10974944 SNP, which might predispose to the acquisition of somatic mutation JAK2(V617F). Acta Haematol. 2010;123:84-7 pubmed publisher
  146. Willmore Payne C, Holden J, Tripp S, Layfield L. Human malignant melanoma: detection of BRAF- and c-kit-activating mutations by high-resolution amplicon melting analysis. Hum Pathol. 2005;36:486-93 pubmed
  147. Karbalaie Niya M, Basi A, Koochak A, Safarnezhad Tameshkel F, Rakhshani N, Zamani F, et al. Sensitive High-Resolution Melting Analysis for Screening of KRAS and BRAF Mutations in Iranian Human Metastatic Colorectal Cancers. Asian Pac J Cancer Prev. 2016;17:5147-5152 pubmed
  148. Illson M, Dempsey Nunez L, Kent J, Huang Q, Brebner A, Raff M, et al. High resolution melting analysis of the MMAB gene in cblB patients and in those with undiagnosed methylmalonic aciduria. Mol Genet Metab. 2013;110:86-9 pubmed publisher
  149. Liu Y, Chiang S, Lin C, Chang J, Chung C, Ko A, et al. Somatic Mutations and Genetic Variants of NOTCH1 in Head and Neck Squamous Cell Carcinoma Occurrence and Development. Sci Rep. 2016;6:24014 pubmed publisher
  150. McConechy M, Anglesio M, Kalloger S, Yang W, Senz J, Chow C, et al. Subtype-specific mutation of PPP2R1A in endometrial and ovarian carcinomas. J Pathol. 2011;223:567-73 pubmed publisher
  151. Dufva M, Poulsen L. Genotyping of mutation in the beta-globin gene using DNA microarrays. Methods Mol Biol. 2009;509:47-56 pubmed publisher
  152. Ozkumur E, Ahn S, Yalçin A, Lopez C, Cevik E, Irani R, et al. Label-free microarray imaging for direct detection of DNA hybridization and single-nucleotide mismatches. Biosens Bioelectron. 2010;25:1789-95 pubmed publisher
  153. Kozlowski P, Jasinska A, Kwiatkowski D. New applications and developments in the use of multiplex ligation-dependent probe amplification. Electrophoresis. 2008;29:4627-36 pubmed publisher
  154. Hömig Hölzel C, Savola S. Multiplex ligation-dependent probe amplification (MLPA) in tumor diagnostics and prognostics. Diagn Mol Pathol. 2012;21:189-206 pubmed publisher
  155. Massalska D, Bijok J, Zimowski J, Jóźwiak A, Jakiel G, Roszkowski T. Multiplex ligation-dependent probe amplification (MLPA)--new possibilities of prenatal diagnosis. Ginekol Pol. 2013;84:461-4 pubmed
  156. Willis A, Van den Veyver I, Eng C. Multiplex ligation-dependent probe amplification (MLPA) and prenatal diagnosis. Prenat Diagn. 2012;32:315-20 pubmed publisher
  157. Grinspan D. [Oral florid papillomatosis or verrucous carcinoma]. Acta Odontol Venez. 1978;16:21-38 pubmed
  158. Nasu M, Emi M, Pastorino S, Tanji M, Powers A, Luk H, et al. High Incidence of Somatic BAP1 alterations in sporadic malignant mesothelioma. J Thorac Oncol. 2015;10:565-76 pubmed publisher
  159. Carnevali I, Libera L, Chiaravalli A, Sahnane N, Furlan D, Viel A, et al. Somatic Testing on Gynecological Cancers Improve the Identification of Lynch Syndrome. Int J Gynecol Cancer. 2017;27:1543-1549 pubmed publisher
  160. Mori T, Sumii M, Fujishima F, Ueno K, Emi M, Nagasaki M, et al. Somatic alteration and depleted nuclear expression of BAP1 in human esophageal squamous cell carcinoma. Cancer Sci. 2015;106:1118-29 pubmed publisher
  161. McKay V, Cairns D, Gokhale D, Mountford R, Greenhalgh L. First report of somatic mosaicism for mutations in STK11 in four patients with Peutz-Jeghers syndrome. Fam Cancer. 2016;15:57-61 pubmed publisher
  162. Grandone A, Del Vecchio Blanco F, Torella A, Caruso M, De Luca F, Di Mase R, et al. Multiplex Ligation-Dependent Probe Amplification Accurately Detects Turner Syndrome in Girls with Short Stature. Horm Res Paediatr. 2016;86:330-336 pubmed publisher
  163. Sanger F, Nicklen S, Coulson A. DNA sequencing with chain-terminating inhibitors. Proc Natl Acad Sci U S A. 1977;74:5463-7 pubmed
  164. Splendore A, Silva E, Alonso L, Richieri Costa A, Alonso N, Rosa A, et al. High mutation detection rate in TCOF1 among Treacher Collins syndrome patients reveals clustering of mutations and 16 novel pathogenic changes. Hum Mutat. 2000;16:315-22 pubmed
  165. Park W, Theda C, Maestri N, Meyers G, Fryburg J, Dufresne C, et al. Analysis of phenotypic features and FGFR2 mutations in Apert syndrome. Am J Hum Genet. 1995;57:321-8 pubmed
  166. Roa I, Garcia H, Game A, De Toro G, de Aretxabala X, Javle M. Somatic Mutations of PI3K in Early and Advanced Gallbladder Cancer: Additional Options for an Orphan Cancer. J Mol Diagn. 2016;18:388-394 pubmed publisher
  167. Wang J, Yang H, Shen Y, Wang S, Lin D, Ma L, et al. Direct sequencing is a reliable assay with good clinical applicability for KRAS mutation testing in colorectal cancer. Cancer Biomark. 2013;13:89-97 pubmed publisher
  168. Albitar A, Ma W, Albitar M. Wild-type Blocking PCR Combined with Direct Sequencing as a Highly Sensitive Method for Detection of Low-Frequency Somatic Mutations. J Vis Exp. 2017;: pubmed publisher
  169. Mardis E. New strategies and emerging technologies for massively parallel sequencing: applications in medical research. Genome Med. 2009;1:40 pubmed publisher
  170. Mardis E. DNA sequencing technologies: 2006-2016. Nat Protoc. 2017;12:213-218 pubmed publisher
  171. Mardis E. Next-generation sequencing platforms. Annu Rev Anal Chem (Palo Alto Calif). 2013;6:287-303 pubmed publisher
  172. Gundry M, Vijg J. Direct mutation analysis by high-throughput sequencing: from germline to low-abundant, somatic variants. Mutat Res. 2012;729:1-15 pubmed publisher
  173. Harding K, Robertson N. Applications of next-generation whole exome sequencing. J Neurol. 2014;261:1244-6 pubmed publisher
  174. Jiang L, Gu Z, Yan Z, Zhao X, Xie Y, Zhang Z, et al. Exome sequencing identifies somatic mutations of DDX3X in natural killer/T-cell lymphoma. Nat Genet. 2015;47:1061-6 pubmed publisher
  175. de Koning T, Jongbloed J, Sikkema Raddatz B, Sinke R. Targeted next-generation sequencing panels for monogenetic disorders in clinical diagnostics: the opportunities and challenges. Expert Rev Mol Diagn. 2015;15:61-70 pubmed publisher
  176. Schmitt M, Kennedy S, Salk J, Fox E, Hiatt J, Loeb L. Detection of ultra-rare mutations by next-generation sequencing. Proc Natl Acad Sci U S A. 2012;109:14508-13 pubmed publisher
  177. Quispe Tintaya W, Gorbacheva T, Lee M, Makhortov S, Popov V, Vijg J, et al. Quantitative detection of low-abundance somatic structural variants in normal cells by high-throughput sequencing. Nat Methods. 2016;13:584-6 pubmed publisher
  178. Coorens T, Treger T, Al Saadi R, Moore L, Tran M, Mitchell T, et al. Embryonal precursors of Wilms tumor. Science. 2019;366:1247-1251 pubmed publisher
  179. Müllauer L. Next generation sequencing: clinical applications in solid tumours. Memo. 2017;10:244-247 pubmed publisher
  180. Bertram L. Next Generation Sequencing in Alzheimer's Disease. Methods Mol Biol. 2016;1303:281-97 pubmed publisher
  181. Møller R, Dahl H, Helbig I. The contribution of next generation sequencing to epilepsy genetics. Expert Rev Mol Diagn. 2015;15:1531-8 pubmed publisher
  182. Ma Y, Shi N, Li M, Chen F, Niu H. Applications of Next-generation Sequencing in Systemic Autoimmune Diseases. Genomics Proteomics Bioinformatics. 2015;13:242-9 pubmed publisher
  183. VASAN N, Razavi P, Johnson J, Shao H, Shah H, Antoine A, et al. Double PIK3CA mutations in cis increase oncogenicity and sensitivity to PI3Kα inhibitors. Science. 2019;366:714-723 pubmed publisher
  184. Sackmann E, Fulton A, Beebe D. The present and future role of microfluidics in biomedical research. Nature. 2014;507:181-9 pubmed publisher
  185. Kovac J, Voldman J. Intuitive, image-based cell sorting using optofluidic cell sorting. Anal Chem. 2007;79:9321-30 pubmed
  186. Landry Z, Giovanonni S, Quake S, Blainey P. Optofluidic cell selection from complex microbial communities for single-genome analysis. Methods Enzymol. 2013;531:61-90 pubmed publisher
  187. Citri A, Pang Z, Sudhof T, Wernig M, Malenka R. Comprehensive qPCR profiling of gene expression in single neuronal cells. Nat Protoc. 2011;7:118-27 pubmed publisher
  188. Frohlich J, Konig H. New techniques for isolation of single prokaryotic cells. FEMS Microbiol Rev. 2000;24:567-72 pubmed
  189. Saliba A, Westermann A, Gorski S, Vogel J. Single-cell RNA-seq: advances and future challenges. Nucleic Acids Res. 2014;42:8845-60 pubmed publisher
  190. Liang J, Cai W, Sun Z. Single-cell sequencing technologies: current and future. J Genet Genomics. 2014;41:513-28 pubmed publisher
  191. Hume D, Donahue R, Fidler I. The therapeutic effect of human recombinant macrophage colony stimulating factor (CSF-1) in experimental murine metastatic melanoma. Lymphokine Res. 1989;8:69-77 pubmed
  192. Kolodziejczyk A, Kim J, Svensson V, Marioni J, Teichmann S. The technology and biology of single-cell RNA sequencing. Mol Cell. 2015;58:610-20 pubmed publisher
  193. Saadatpour A, Lai S, Guo G, Yuan G. Single-Cell Analysis in Cancer Genomics. Trends Genet. 2015;31:576-86 pubmed publisher
  194. Yu M, Stott S, Toner M, Maheswaran S, Haber D. Circulating tumor cells: approaches to isolation and characterization. J Cell Biol. 2011;192:373-82 pubmed publisher
  195. Altomare L, Borgatti M, Medoro G, Manaresi N, Tartagni M, Guerrieri R, et al. Levitation and movement of human tumor cells using a printed circuit board device based on software-controlled dielectrophoresis. Biotechnol Bioeng. 2003;82:474-9 pubmed
  196. De Luca F, Rotunno G, Salvianti F, Galardi F, Pestrin M, Gabellini S, et al. Mutational analysis of single circulating tumor cells by next generation sequencing in metastatic breast cancer. Oncotarget. 2016;7:26107-19 pubmed publisher
  197. Fernandez S, Bingham C, Fittipaldi P, Austin L, Palazzo J, Palmer G, et al. TP53 mutations detected in circulating tumor cells present in the blood of metastatic triple negative breast cancer patients. Breast Cancer Res. 2014;16:445 pubmed publisher
  198. Pestrin M, Salvianti F, Galardi F, De Luca F, Turner N, Malorni L, et al. Heterogeneity of PIK3CA mutational status at the single cell level in circulating tumor cells from metastatic breast cancer patients. Mol Oncol. 2015;9:749-57 pubmed publisher
  199. Paolillo C, Mu Z, Rossi G, Schiewer M, Nguyen T, Austin L, et al. Detection of Activating Estrogen Receptor Gene (ESR1) Mutations in Single Circulating Tumor Cells. Clin Cancer Res. 2017;23:6086-6093 pubmed publisher
  200. Sho S, Court C, Winograd P, Lee S, Hou S, Graeber T, et al. Precision oncology using a limited number of cells: optimization of whole genome amplification products for sequencing applications. BMC Cancer. 2017;17:457 pubmed publisher
  201. Jiang R, Lu Y, Ho H, Li B, Chen J, Lin M, et al. A comparison of isolated circulating tumor cells and tissue biopsies using whole-genome sequencing in prostate cancer. Oncotarget. 2015;6:44781-93 pubmed publisher
  202. Court C, Ankeny J, Sho S, Hou S, Li Q, Hsieh C, et al. Reality of Single Circulating Tumor Cell Sequencing for Molecular Diagnostics in Pancreatic Cancer. J Mol Diagn. 2016;18:688-696 pubmed publisher
  203. Huang L, Ma F, Chapman A, Lu S, Xie X. Single-Cell Whole-Genome Amplification and Sequencing: Methodology and Applications. Annu Rev Genomics Hum Genet. 2015;16:79-102 pubmed publisher
  204. Palmirotta R, Lovero D, Silvestris E, Felici C, Quaresmini D, Cafforio P, et al. Next-generation Sequencing (NGS) Analysis on Single Circulating Tumor Cells (CTCs) with No Need of Whole-genome Amplification (WGA). Cancer Genomics Proteomics. 2017;14:173-179 pubmed
  205. Powell A, Talasaz A, Zhang H, Coram M, Reddy A, Deng G, et al. Single cell profiling of circulating tumor cells: transcriptional heterogeneity and diversity from breast cancer cell lines. PLoS ONE. 2012;7:e33788 pubmed publisher
  206. Deng G, Krishnakumar S, Powell A, Zhang H, Mindrinos M, Telli M, et al. Single cell mutational analysis of PIK3CA in circulating tumor cells and metastases in breast cancer reveals heterogeneity, discordance, and mutation persistence in cultured disseminated tumor cells from bone marrow. BMC Cancer. 2014;14:456 pubmed publisher
  207. Xu X, Zhu K, Liu F, Wang Y, Shen J, Jin J, et al. Identification of somatic mutations in human prostate cancer by RNA-Seq. Gene. 2013;519:343-7 pubmed publisher
  208. Sheng Q, Zhao S, Li C, Shyr Y, Guo Y. Practicability of detecting somatic point mutation from RNA high throughput sequencing data. Genomics. 2016;107:163-9 pubmed publisher
  209. . Comprehensive molecular portraits of human breast tumours. Nature. 2012;490:61-70 pubmed publisher
  210. Lin W, Piskol R, Tan M, Li J. Comment on "Widespread RNA and DNA sequence differences in the human transcriptome". Science. 2012;335:1302; author reply 1302 pubmed publisher
  211. Pickrell J, Gilad Y, Pritchard J. Comment on "Widespread RNA and DNA sequence differences in the human transcriptome". Science. 2012;335:1302; author reply 1302 pubmed publisher
  212. Guo Y, Li J, Li C, Long J, Samuels D, Shyr Y. The effect of strand bias in Illumina short-read sequencing data. BMC Genomics. 2012;13:666 pubmed publisher
  213. Piskol R, Ramaswami G, Li J. Reliable identification of genomic variants from RNA-seq data. Am J Hum Genet. 2013;93:641-51 pubmed publisher
  214. Duitama J, Srivastava P, Mandoiu I. Towards accurate detection and genotyping of expressed variants from whole transcriptome sequencing data. BMC Genomics. 2012;13 Suppl 2:S6 pubmed publisher
  215. Radenbaugh A, Ma S, Ewing A, Stuart J, Collisson E, Zhu J, et al. RADIA: RNA and DNA integrated analysis for somatic mutation detection. PLoS ONE. 2014;9:e111516 pubmed publisher
  216. Hansen M, Herborg L, Hansen M, Roug A, Hokland P. Combination of RNA- and exome sequencing: Increasing specificity for identification of somatic point mutations and indels in acute leukaemia. Leuk Res. 2016;51:27-31 pubmed publisher
  217. Karasaki T, Nagayama K, Kuwano H, Nitadori J, Sato M, Anraku M, et al. Prediction and prioritization of neoantigens: integration of RNA sequencing data with whole-exome sequencing. Cancer Sci. 2017;108:170-177 pubmed publisher
  218. Cibulskis K, Lawrence M, Carter S, Sivachenko A, Jaffe D, Sougnez C, et al. Sensitive detection of somatic point mutations in impure and heterogeneous cancer samples. Nat Biotechnol. 2013;31:213-9 pubmed publisher
  219. Yizhak K, Aguet F, Kim J, Hess J, Kübler K, Grimsby J, et al. RNA sequence analysis reveals macroscopic somatic clonal expansion across normal tissues. Science. 2019;364: pubmed publisher
  220. Watson I, Takahashi K, Futreal P, Chin L. Emerging patterns of somatic mutations in cancer. Nat Rev Genet. 2013;14:703-18 pubmed publisher
  221. Jones S, Anagnostou V, Lytle K, Parpart Li S, Nesselbush M, Riley D, et al. Personalized genomic analyses for cancer mutation discovery and interpretation. Sci Transl Med. 2015;7:283ra53 pubmed publisher
  222. McCarthy M. Genomic sequencing of only tumor tissue could be misleading in nearly half of patients, study shows. BMJ. 2015;350:h2036 pubmed publisher
  223. Teer J, Zhang Y, Chen L, Welsh E, Cress W, Eschrich S, et al. Evaluating somatic tumor mutation detection without matched normal samples. Hum Genomics. 2017;11:22 pubmed publisher
  224. McKenna A, Hanna M, Banks E, Sivachenko A, Cibulskis K, Kernytsky A, et al. The Genome Analysis Toolkit: a MapReduce framework for analyzing next-generation DNA sequencing data. Genome Res. 2010;20:1297-303 pubmed publisher
ISSN : 2329-5139