Fact-checked by Grok 2 weeks ago

Variants of PCR

(PCR) is a fundamental technique invented by in 1983 that enables the exponential amplification of specific DNA sequences through repeated cycles of denaturation, annealing, and extension using a thermostable , such as derived from . Variants of PCR represent specialized adaptations of this core method, designed to overcome limitations of the original protocol—such as sensitivity to contamination, inability to quantify products in real time, or challenges with RNA templates—and to expand its utility across diverse applications including diagnostics, forensics, and . These modifications incorporate innovations like fluorescent probes, multiple primer sets, or sample partitioning to improve specificity, enable , or provide absolute quantification without reliance on standard curves. Among the most notable variants is real-time quantitative PCR (qPCR), which integrates fluorescence detection during amplification to monitor reaction progress in real time, allowing for precise quantification of DNA or RNA targets and serving as the gold standard for applications like viral load assessment in infectious disease diagnostics, such as SARS-CoV-2 detection. Reverse transcription PCR (RT-PCR) extends the technique to RNA by first converting it to complementary DNA (cDNA) via reverse transcriptase, facilitating gene expression analysis and RNA virus identification, with widespread use in clinical settings for rapid pathogen detection. Multiplex PCR amplifies multiple DNA targets simultaneously in a single reaction using distinct primer pairs, enhancing efficiency for tasks like forensic DNA profiling or simultaneous screening of genetic mutations in hereditary diseases. Further advancements include digital PCR (dPCR), which partitions the sample into thousands or millions of microreactions—via droplets (ddPCR) or chips (cdPCR)—to achieve absolute quantification by counting positive partitions, offering superior precision for rare mutation detection and analysis in . Nested PCR employs two sequential amplification rounds with inner and outer primer sets to boost specificity and reduce non-specific products, particularly useful in low-abundance template scenarios like studies. Techniques such as hot-start PCR, which activates the polymerase only at high temperatures to minimize primer-dimer formation, and touchdown PCR, which progressively lowers annealing temperatures for optimized specificity, address common artifacts in standard . Emerging isothermal variants, like (LAMP), eliminate thermal cycling altogether for in resource-limited environments. Collectively, these variants have transformed into a versatile toolkit, underpinning next-generation sequencing library preparation and single-cell analyses while driving innovations in portable, microfluidic devices for on-site diagnostics.

Cycle and Protocol Modifications

Hot-start PCR

Hot-start PCR is a modification of the standard () designed to minimize non-specific amplification by inhibiting activity at ambient temperatures during reaction setup, thereby preventing primer-dimer formation and mispriming. This technique ensures that polymerase activation occurs only after the initial high-temperature denaturation step, typically at 95°C, enhancing the specificity and yield of the target amplicon. By addressing the intrinsic activity of Taq at , hot-start PCR has become essential for applications requiring high precision. The technique was introduced in the late 1980s to overcome limitations in early protocols, where non-specific products compromised results, particularly in diagnostic and assays. Early implementations focused on manual addition of post-denaturation, but automated methods emerged in the , including the use of inhibitory agents that are released upon heating. These developments were driven by the need to improve in high-throughput settings, with commercial hot-start enzymes like AmpliTaq becoming widely adopted by the early . Activation methods for hot-start PCR fall into three main categories: chemical, enzymatic, and physical. Chemical inhibition involves modifying the with groups, such as in FastStart Taq , where blocking groups are removed during a 2-minute at 95°C to activate the . Enzymatic methods use reversible inhibitors like antibodies or aptamers; for example, Taq employs antibodies that dissociate at 70°C, while AptaTaq uses DNA aptamers that release above 70°C for faster activation. Physical methods rely on barriers to separate components until melting at high temperatures, such as wax beads that form a seal between polymerase and primers/dNTPs, or dried trehalose-embedded beads for small-volume reactions. These approaches allow room-temperature assembly while ensuring inactivation until the activation step, typically 2-10 minutes at 95°C depending on the method. In routine diagnostics and , hot-start PCR is particularly valuable for amplifying low-abundance targets in complex samples, such as viral detection or , where specificity is paramount. It is often combined with nested PCR for enhanced multi-step specificity in challenging templates. The primary advantages include a significant reduction in background noise from non-specific products—often by orders of magnitude—leading to cleaner gels and higher target yields, alongside improved sensitivity in quantitative assays.

Touchdown PCR

Touchdown PCR is a modification of the standard () designed to improve the specificity of amplification by progressively lowering the annealing temperature during the initial cycles, thereby minimizing non-specific priming while maximizing the yield of the target product. This technique was developed in 1991 by R.H. Don and colleagues at the to address challenges in amplifying specific genes from complex genomic templates, where spurious products often outcompete the desired amplicon due to their smaller size and faster replication kinetics. The protocol for touchdown PCR typically begins with an initial denaturation step, followed by 5–10 cycles where the annealing temperature is set 5–10°C above the calculated melting temperature (Tm) of the primers. In these early cycles, the temperature is decreased by 0.5–1°C per cycle (or every second cycle in some variants) until it reaches the standard Tm or 2–5°C below it, after which 20–30 additional cycles are performed at this final annealing temperature, with denaturation at 94–95°C and extension at 72°C. This stepwise reduction ensures that specific primers, which have a higher Tm, anneal preferentially during the high-temperature phase, while non-specific primers with lower Tm are less likely to bind. The mechanism relies on the exponential nature of : in the initial high-stringency , correct primer-template hybrids are favored, providing them with a kinetic advantage of approximately 2-fold per (or 4-fold per °C difference in ) over mismatched annealings, which suppresses off-target products before they can accumulate. As the "touches down" to optimal levels, of the specific product proceeds efficiently without significant from non-specific bands. Touchdown PCR is particularly useful for amplifying low-abundance targets in genomic DNA from complex samples, such as in pathogen detection where it enables sensitive identification of bacterial or protozoan DNA in environmental or clinical matrices. It has also been applied in ecological studies, including environmental DNA (eDNA) analysis for tracking microbial or wildlife populations in aquatic and soil ecosystems. For maximal specificity, touchdown PCR is often combined with hot-start PCR techniques to further prevent primer-dimer formation. Optimization of touchdown PCR involves adjusting the total temperature decrease to 10–20°C over the initial cycles, with the number of touchdown cycles (typically 10–15) tailored to template complexity—more cycles for highly complex or low-input DNA to build specificity early. This approach reduces the need for empirical testing of fixed annealing temperatures and enhances reproducibility across different thermal cyclers.

Nested PCR

Nested PCR is a modification of the standard () designed to improve both specificity and , particularly for amplifying rare or low-abundance target sequences in complex samples. The technique involves two sequential amplification steps: in the first round, a pair of outer primers is used to generate an intermediate-length product from the target DNA template over 15–30 cycles. An aliquot of this product (typically 1–10% of the reaction volume) then serves as the template for the second round, where a pair of inner primers—located internally within the outer primer binding sites—amplifies a shorter, more specific fragment over an additional 25–35 cycles. This nested approach reduces non-specific amplification by minimizing the chances of primer mispriming in the initial broad amplification, as the inner primers only bind to the correctly amplified intermediate product. The method was first introduced in 1988 by Saiki et al. for detecting human immunodeficiency virus (HIV) in clinical samples, marking an early advancement in PCR-based viral diagnostics. By combining the thermostable Taq polymerase with this two-step primer strategy, nested PCR achieves a sensitivity gain of up to 10^4-fold compared to conventional assays like slot-blot hybridization, enabling reliable detection of as few as 1–10 target template molecules in a background of excess non-target DNA. One study demonstrated this enhanced detection for hepatitis B virus serum DNA, where nested PCR identified positives in samples negative by standard methods, underscoring its utility for low-copy viral targets. A primary challenge with nested PCR is the risk of carryover contamination, where amplicons from the first round aerosolize and template false positives in the second round or subsequent reactions. This is mitigated by incorporating deoxyuridine triphosphate (dUTP) in place of dTTP during amplification, followed by treatment with uracil-DNA glycosylase (UDG) to selectively degrade uracil-containing contaminant DNA prior to the second round, while preserving the native template. Nested PCR finds key applications in forensics, where it amplifies trace DNA from mixed or degraded evidence like on surfaces, and in ancient DNA analysis, allowing recovery of genetic material from millennia-old samples with minimal starting material. Variations include fully nested PCR, which employs entirely new inner primers for maximum specificity, and semi-nested PCR, which shares one primer from the outer set with a new internal primer to streamline the process while retaining much of the sensitivity benefit. In quantification contexts, techniques like digital PCR can sometimes obviate the need for nested amplification by partitioning samples into thousands of reactions for absolute counting of rare targets.

Asymmetric PCR

Asymmetric PCR is a modification of the standard () that employs unequal concentrations of the two primers to preferentially amplify one strand of the target DNA, resulting in the production of single-stranded DNA (ssDNA) alongside double-stranded DNA (dsDNA). This technique was first described in by Gyllensten and Erlich as a method to generate ssDNA suitable for direct sequencing of PCR products, eliminating the need for steps. The process involves an initial exponential amplification phase where both primers are utilized to produce dsDNA, followed by a linear amplification phase after depletion of the limiting primer, during which the excess primer extends the ssDNA template repeatedly. In the protocol, primers are typically added in a ratio of 50:1 to 100:1 (limiting primer to excess primer), allowing the reaction to shift from balanced dsDNA synthesis to unbalanced ssDNA production. For example, the excess primer is often used at concentrations of 20-50 nM, while the limiting primer is at 0.2-1 nM, though adjustments may be needed based on template and amplicon length. The reaction proceeds through standard thermal cycling (denaturation, annealing, extension) for 30-50 cycles, with the linear phase yielding 10-100 times more ssDNA than dsDNA under optimized conditions. Post-PCR cleanup, such as treatment or gel purification, is commonly required to isolate the desired ssDNA by removing excess primers and dsDNA contaminants. This variant finds applications in generating ssDNA probes for hybridization assays, where the single-stranded nature enhances binding efficiency to target sequences in microarrays or northern blots. It is also employed in direct sequencing of PCR amplicons without , as the enriched ssDNA serves as an ideal template for . Additionally, asymmetric PCR supports probe-based detection in diagnostic assays, leveraging the ssDNA for improved specificity in hybridization reactions. Optimization of asymmetric PCR focuses on primer ratio and concentration to maximize ssDNA yield while minimizing non-specific products, often requiring empirical testing for each target. However, the method generally produces lower overall yields compared to symmetric PCR due to the transition to linear amplification, and imbalances can introduce artifacts such as primer-dimers or off-target amplification.

Quantitative and Detection Variants

Real-time PCR (qPCR)

Real-time , also known as quantitative (qPCR), enables the monitoring and quantification of DNA in real time by incorporating fluorescent detection directly into the cycles, eliminating the need for post- analysis such as . This technique was first developed in by Higuchi et al., who demonstrated the use of to track product accumulation via video imaging during thermal cycling. By measuring at each cycle, qPCR provides kinetic data on , allowing for precise determination of initial template quantities based on the exponential phase of the reaction. Detection in qPCR relies on two primary methods: intercalating dyes and sequence-specific probes. Intercalating dyes, such as , bind nonspecifically to double-stranded DNA (dsDNA) produced during amplification, resulting in increased fluorescence proportional to amplicon accumulation; this approach is cost-effective but can detect nonspecific products, necessitating melt curve analysis for specificity. In contrast, probe-based methods like utilize fluorogenic probes labeled with a reporter at the 5' end and a quencher at the 3' end; during extension, the 5' activity of cleaves the probe, separating the reporter from the quencher and generating a detectable signal specific to the target sequence. This probe mechanism, originally described by Holland et al. in 1991, enhances specificity and enables through probes with distinct fluorophores. Quantification in qPCR is achieved through the cycle threshold (Ct) value, defined as the PCR cycle number at which the fluorescence signal crosses a predetermined threshold above background noise, reflecting the point of exponential amplification. For absolute quantification, a standard curve is generated by plotting Ct values against known template concentrations, from which amplification efficiency (E) is calculated using the formula E = 10^{-1/\text{slope}}; optimal efficiencies range from 90% to 110%, corresponding to slopes of -3.58 to -3.10. Relative quantification, commonly used for gene expression analysis, employs the ΔΔCt method, which compares the difference in Ct values (ΔCt) between target and reference genes across samples, normalized to a calibrator; this approach assumes similar efficiencies between targets. Expression levels are often normalized to housekeeping genes such as GAPDH to account for variations in input RNA or DNA. qPCR finds widespread applications in , where it quantifies mRNA levels by first performing reverse transcription to generate cDNA (RT-qPCR); viral load monitoring in clinical diagnostics, such as or quantification; and (SNP) genotyping through allele-specific probes. These uses leverage qPCR's high sensitivity, detecting as few as 10 copies of target , and its spanning five to six orders of magnitude. Instruments for qPCR consist of modified thermocyclers integrated with optical systems for fluorescence detection, typically supporting 4-5 color channels for up to 2-5 targets simultaneously, limited by overlap and design complexity. Advanced systems, such as those from , incorporate multiple excitation sources and detectors to minimize crosstalk in multiplex reactions.

Digital PCR

PCR (dPCR) is a method for absolute quantification that partitions a sample into thousands to millions of individual microreactions, each containing zero or few target molecules, enabling direct counting of positive partitions after endpoint amplification. This approach relies on statistics to estimate target concentration without requiring standard curves or amplification efficiency assumptions, distinguishing it from relative quantification methods like qPCR. The technique was first conceptualized in the early 1990s through pioneering work on limiting dilution and for target quantitation. In droplet digital PCR (ddPCR), the most widely adopted format, the sample is emulsified into 10,000 to 100,000 oil-encapsulated aqueous droplets, with each droplet serving as an independent reaction volume of approximately 1 nanoliter. After thermal cycling, droplets are analyzed for to classify them as positive (containing amplified target) or negative, using a droplet reader that employs principles to stream and detect individual droplets at high throughput. The average number of target molecules per partition, denoted as λ, is calculated via the formula: \lambda = -\ln(1 - p) where p represents the fraction of positive droplets. The total target copy number is then determined as λ multiplied by the total number of partitions and adjusted for any dilution factor, providing precise absolute quantification with high reproducibility. This method was commercialized in 2011 by Bio-Rad Laboratories following their acquisition of QuantaLife, building on foundational ddPCR demonstrations. dPCR excels in applications requiring high sensitivity, such as detecting copy number variations (CNVs) in genomic DNA, where it can resolve differences as small as 10% without reference standards, and rare mutation detection in heterogeneous samples like circulating tumor DNA, achieving sensitivities down to 0.01% variant allele frequency. Its advantages include robust performance in the presence of PCR inhibitors—such as humic acids or heme from clinical samples—due to the isolation of reactions in partitions, which minimizes global inhibition effects, and reliance on endpoint readouts rather than real-time kinetics, eliminating biases from variable amplification efficiencies. Unlike qPCR, dPCR does not assume perfect doubling per cycle, yielding more accurate results for low-abundance targets. Variations of dPCR include droplet-based systems like Bio-Rad's QX200, which generate flexible partitions, and chamber-based platforms such as Thermo Fisher's QuantStudio 3D, which use preloaded microfluidic chips to form fixed arrays of up to 20,000 wells for more consistent partition volumes and simplified workflows. Both formats leverage Poisson-based analysis but differ in throughput and ease of use, with droplet methods offering higher partition numbers for enhanced precision in ultra-low target detection.

COLD-PCR

COLD-PCR, or co-amplification at lower denaturation PCR, is a modification of standard designed to selectively enrich low-abundance DNA alleles from a background of wild-type DNA. The method exploits the principle that heteroduplexes formed between wild-type and mutant strands have a lower (T_m) due to sequence mismatches, allowing for preferential denaturation of these heteroduplexes at a critical (T_c) that is typically 1-2°C below the full T_m of the amplicon. This enables the to preferentially amplify single-stranded templates while reannealing wild-type strands, resulting in exponential enrichment of variants without requiring specialized primers or enzymes. The protocol for COLD-PCR involves an initial phase of conventional PCR cycles (usually 5-10) at full denaturation temperature (e.g., 94-95°C) to generate amplicons, followed by alternating full and partial denaturation cycles at T_c (e.g., 80-85°C, optimized computationally or empirically for each sequence). Annealing and extension steps occur at standard temperatures (50-60°C and 72°C, respectively), with cycle numbers tailored to achieve desired enrichment (typically 20-40 cycles total). This approach can yield up to 100-fold enrichment of mutant alleles, enabling detection of variants at frequencies as low as 0.1-1% in heterogeneous samples. T_c selection is crucial, as it must dissociate mutant-containing heteroduplexes while preserving perfect-match wild-type duplexes. Developed in 2008 by Li et al. as a tool to enhance mutation detection sensitivity in clinical settings, COLD-PCR has been particularly applied in cancer diagnostics for identifying somatic mutations in genes like KRAS from tumor biopsies or circulating DNA. For instance, in colorectal and other solid tumors, it has improved KRAS mutation detection in formalin-fixed paraffin-embedded samples by over 4-fold compared to conventional PCR, facilitating personalized therapy decisions such as anti-EGFR treatment eligibility. The technique's specificity stems from its reliance on sequence-specific mismatches rather than probe hybridization, making it broadly applicable to point mutations, insertions, deletions, and methylation events; it is often combined with downstream sequencing (e.g., Sanger or next-generation) for validation. Several variants of COLD-PCR address limitations in mutation type specificity and efficiency. Fast-COLD-PCR employs a simplified three-step cycling (denaturation at T_c, annealing/extension) without a separate hybridization step, using rapid cooling (e.g., via ) to promote heteroduplex formation; it is optimized for Tm-reducing s and achieves ~20-fold enrichment but is faster and suitable for high-throughput assays. Ice-COLD-PCR, introduced in by Milbury et al., incorporates a non-extendable wild-type reference added post-initial to force heteroduplex formation with mutants, enabling robust enrichment (up to 75-fold) for all mutation types, including Tm-neutral or Tm-increasing ones, through enhanced strand displacement during the T_c step. These variants maintain compatibility with standard thermal cyclers and have expanded COLD-PCR's utility in detecting rare alleles in liquid biopsies.

RNA and Pretreatment Variants

Reverse-transcription PCR (RT-PCR)

Reverse-transcription PCR (RT-PCR) is a laboratory technique that enables the amplification and analysis of molecules by first synthesizing (cDNA) from RNA templates using , followed by amplification of the cDNA. This variant addresses the limitation of standard , which amplifies only DNA, making RT-PCR essential for studying RNA viruses, , and transcriptomics. The method was first described in 1987 by Kawasaki et al., building on the earlier invention of in 1983 and in 1970. The procedure begins with reverse transcription, where an RNA-dependent DNA polymerase, such as Moloney murine leukemia virus (M-MLV) reverse transcriptase, synthesizes cDNA from the RNA template. M-MLV RT typically includes RNase H activity, which cleaves the RNA strand in the RNA-DNA hybrid after cDNA synthesis, facilitating processivity. Primers for this step include oligo(dT) primers that bind to the poly-A tail of mRNA, random hexamers for unbiased synthesis from total RNA, or gene-specific primers for targeted amplification. The reaction is optimized at 42–50°C incubation to reduce RNA secondary structures that could impede synthesis, with durations of 30–60 minutes depending on template length. RT-PCR protocols are categorized as one-step, where reverse transcription and PCR occur in a single tube using a thermostable reverse transcriptase like Tth polymerase to minimize handling and contamination, or two-step, where cDNA is synthesized separately and then aliquoted for multiple PCR reactions, offering greater flexibility for downstream applications. RT-PCR finds widespread use in to quantify mRNA abundance across tissues or conditions, and in viral RNA detection, notably for pathogens like where it serves as the gold standard for diagnostic testing by targeting viral genes such as the nucleocapsid or . For quantitative applications, RT-qPCR integrates fluorescence detection during to measure mRNA levels accurately, achieving down to 10–100 RNA molecules per reaction, which is critical for low-abundance transcripts. A primary challenge is RNA degradation by environmental RNases, mitigated by incorporating inhibitors like RNasin, a recombinant inhibitor that binds and neutralizes RNases without interfering with the enzymatic reactions. Proper handling, including RNase-free reagents and conditions, ensures reliable results.

Methylation-specific PCR

Methylation-specific () is a technique designed to detect and distinguish between methylated and unmethylated residues in CpG dinucleotides, particularly within CpG islands of promoters. The method relies on treatment of genomic DNA, which deaminates unmethylated cytosines to uracils (that are read as thymines during ), while 5-methylcytosines remain unchanged. Following treatment, amplification uses primers that are sequence-specific: those targeting unmethylated alleles incorporate thymines at former cytosine positions, whereas primers for methylated alleles match the original guanine- pairing. This allows qualitative assessment of status at specific loci using standard conditions, typically involving 35 cycles of denaturation at 95°C, annealing, and extension at 72°C, followed by . Developed in 1996 by Herman et al., has become a cornerstone in research, enabling the analysis of patterns associated with . It requires minimal DNA input (as low as 50 ng) and is applicable to archived samples like paraffin-embedded tissues. In cancer detection, MSP has identified promoter hypermethylation in tumor suppressor genes, such as the gene in , where methylated promoters predict improved survival with chemotherapy (median survival 21.7 months versus 15.3 months without methylation). For instance, in a of 206 glioblastoma patients, 45% showed MGMT promoter methylation via MSP, correlating with therapeutic response. The technique's sensitivity detects as little as 0.1% methylated alleles in a background of unmethylated DNA, making it suitable for heterogeneous tumor samples. Despite its advantages, MSP faces limitations including incomplete bisulfite conversion, which can lead to false positives by leaving unmethylated cytosines undetected, and DNA degradation during the 16-hour treatment, potentially reducing yield by over 75%. These issues are mitigated by optimized protocols using commercial kits with shorter incubation times. For , methylation-specific (MS-qPCR), also known as MethyLight, incorporates real-time fluorescence detection with probes to measure ratios accurately across a . A variant, HeavyMethyl PCR, enhances specificity for heavily methylated regions by using methylation-specific blockers to suppress unmethylated amplification, combined with probes for real-time quantification; it is particularly useful in clinical settings for low-input samples. Validation of MSP results often involves , which provides semi-quantitative confirmation of levels at individual CpG sites.

Ligation-mediated PCR

Ligation-mediated PCR (LM-PCR) is a technique that enables the amplification of unknown DNA sequences adjacent to a known region by ligating universal linkers or adapters to the ends of fragmented genomic DNA, allowing subsequent PCR amplification using one gene-specific primer and one linker-specific primer. This method is particularly useful for genome walking, where it facilitates the step-wise extension into flanking regions without prior knowledge of the sequence. The procedure involves first digesting genomic DNA with a to generate fragments with blunt or compatible ends, followed by blunt-end of a universal linker using to create a common priming site on the unknown ends. Subsequent amplification employs a gene-specific primer targeting the known and a primer annealing to the ligated linker, often in a nested format to enhance specificity and suppress non-specific products by requiring two rounds of amplification with internal primers. High-quality is essential, typically achieved with under optimized conditions to ensure efficient adapter attachment and minimize chimeric products. Developed in 1989 by Paul R. Mueller and Barbara Wold for footprinting and genomic sequencing, LM-PCR has been adapted for applications such as transposon tagging, where it identifies insertion sites by amplifying flanking genomic sequences post-transposon . It is also widely employed in mapping sites, particularly for , by selectively amplifying junctions between and host DNA. A notable variation is (MLPA), introduced in , which uses pairs of probes that ligate only upon hybridization to target sequences for quantitative detection of copy number variations across multiple loci in a single reaction. MLPA suppresses non-specific amplification through probe design and nested linker strategies, enabling reliable quantification without . In terms of efficiency, LM-PCR typically allows genome walking of 50-500 base pairs per reaction, depending on fragment size and primer design, making it suitable for targeted extension but often requiring iterative rounds for longer distances. Modern implementations frequently integrate LM-PCR with next-generation sequencing (NGS) for high-throughput analysis, such as parallel mapping of thousands of integration sites in viral studies. Unlike inverse PCR, which relies on circularization of templates for outward priming from known sequences, LM-PCR uses linear linker for open-ended exploration of adjacent regions.

Primer and Specificity Modifications

Allele-specific PCR

Allele-specific PCR (AS-PCR), also referred to as the amplification refractory mutation system (), is a PCR variant that enables the selective of specific alleles or genetic polymorphisms through the use of primers designed to anneal exclusively to the target sequence. This technique exploits the inability of DNA polymerases to efficiently extend primers with a mismatch at the 3' terminus, particularly when the polymorphism, such as a single nucleotide polymorphism (), is positioned at the primer's 3' end. For the non-target , this mismatch destabilizes primer binding and inhibits extension, while on the target allows robust . To further enhance discrimination, incorporates one or more deliberate mismatches in the penultimate or antepenultimate positions from the 3' end of the primer, which amplifies the destabilizing effect of the terminal mismatch and reduces non-specific . Developed in 1989 by et al., AS-PCR has been widely adopted for direct without the need for hybridization probes. Its applications span , where it identifies variants like *4 influencing drug metabolism and response to therapies such as clopidogrel, and disease association studies, including HLA typing for assessing transplant compatibility and autoimmune risk.00138-4/fulltext) Optimization of AS-PCR conditions is crucial for achieving high specificity and . Adjusting Mg²⁺ concentrations, typically in the range of 1.5–2.5 mM, balances primer annealing stability and activity to favor target-specific extension while suppressing mismatches. annealing protocols, which start with higher s (e.g., 5–10°C above the primer ) and gradually decrease them over initial cycles, further improve specificity by promoting selective primer early in the . With these optimizations, AS-PCR can reliably detect target alleles at frequencies as low as 1% in heterogeneous samples, making it suitable for identifying minor variants in clinical settings. Multiplexing in AS-PCR facilitates the simultaneous genotyping of multiple alleles in a single reaction, with protocols supporting up to 10 alleles through meticulous primer design to ensure distinct annealing temperatures, minimal dimer formation, and appropriate product sizes for resolution on agarose gels. This approach requires careful spacing of primers to avoid cross-interference and can integrate with broader multiplex PCR strategies for comprehensive polymorphism screening. Validation of AS-PCR results is commonly performed via Sanger sequencing to confirm allele identity or restriction digest analysis to verify specific amplicon patterns, ensuring accuracy in downstream applications.

Multiplex PCR

Multiplex PCR is a variant of the () that enables the simultaneous amplification of multiple specific DNA target sequences in a single reaction tube using several pairs of primers. This approach increases throughput and reduces time and reagent costs compared to performing separate reactions for each target, making it valuable for applications requiring parallel analysis of genetic markers. The technique was first developed in 1988 by Chamberlain et al. for screening deletions in the locus, where multiple exons were amplified concurrently to detect genetic abnormalities. In designing multiplex PCR assays, typically 3 to 20 primer pairs are selected to target non-overlapping amplicons, each ranging from 100 to 500 base pairs in length, ensuring resolvability by downstream methods such as . Primers are optimized for uniform annealing temperatures, generally around 55–60°C, to promote balanced amplification across all targets and minimize bias. Key applications include diagnostics, such as multiplex panels for detecting sexually transmitted infections like and , and forensics, where short tandem repeat (STR) profiling amplifies multiple loci for human identification. Challenges in multiplex PCR arise primarily from inter-primer interactions and competition for reagents, which can lead to preferential amplification of certain targets and reduced specificity. These issues are often addressed by adjusting primer concentrations to equalize yields and employing touchdown PCR protocols, which start with higher annealing temperatures and gradually decrease them to enhance specificity. Modern commercial kits support high-plex reactions, up to 96 targets or more, through advanced primer design algorithms that mitigate dimer formation and off-target amplification. Detection of multiplex PCR products typically occurs post-amplification via for size separation, for high-resolution , or integration with quantitative real-time for simultaneous amplification and monitoring. However, limitations include overall reduced yield per target due to resource competition and heightened sensitivity to PCR inhibitors, such as or humic acids, which can disproportionately affect multi-target reactions compared to simplex .

Inverse PCR

Inverse PCR (IPCR) is a variant of the polymerase chain reaction designed to amplify unknown DNA sequences flanking a region of known sequence, such as insertion sites or junctions. The method involves digesting genomic DNA with a restriction enzyme to generate fragments containing the known sequence, followed by self-ligation to circularize the DNA, and subsequent PCR amplification using primers oriented outward from the known region toward the unknown flanks. This approach allows symmetric amplification of sequences on both sides of the target, enabling the identification of insertions, deletions, or integrations without prior knowledge of the flanking regions. Developed by Howard Ochman and colleagues in 1988, IPCR was first demonstrated by amplifying sequences flanking an IS1 transposable element in Escherichia coli. The protocol begins with a using an that cuts outside the known , ideally producing fragments of 1-5 to facilitate efficient circularization and . The digested DNA is then ligated under dilute conditions to promote intramolecular joining, forming circular templates. is performed with primers annealing to the known but facing away from it, extending through the unknown regions and across the ligation junction. To enhance specificity and yield, a partial digest may be used to generate a population of fragments with varying distances from the known , and nested primers can be employed in a second round of . IPCR has been widely applied in , including transposon mapping to identify insertion sites in bacterial genomes, analysis of integration sites in host DNA, and characterization of disruptions caused by . For instance, it has been used to map transposon insertions in bacterial chromosomes by amplifying flanking sequences for sequencing. In studies, IPCR facilitates the detection of events, such as those of retroviruses or , by revealing adjacent host sequences. Similarly, it aids in disruption analysis by cloning junctions of inserted elements in model organisms like Dictyostelium. Key advantages of IPCR include its ability to amplify both flanking regions simultaneously in a single reaction, providing balanced coverage that is useful for detecting symmetric insertions or deletions, and its reliance on standard reagents without needing specialized enzymes. As a variation, bisulfite-treated IPCR combines the method with conversion to analyze status in unknown flanking regions, such as those adjacent to integrated proviruses, by converting unmethylated cytosines to uracils prior to circularization and amplification. This adaptation is particularly valuable in for studying patterns at integration sites. Ligation-mediated PCR offers a non-circular alternative using linkers for flanking amplification.

Enzyme and Fidelity Enhancements

High-fidelity PCR

High-fidelity refers to variants of the that employ DNA polymerases to achieve significantly lower error rates during amplification, enabling the production of accurate DNA amplicons, particularly for long sequences. These polymerases possess 3'-5' activity, which allows them to excise and correct mismatched incorporated during synthesis, contrasting with non- enzymes like Taq that lack this capability. Developed to address the limitations of standard in applications requiring precise replication, high-fidelity minimizes mutations, making it essential for downstream processes where sequence integrity is paramount. The seminal high-fidelity polymerase, Pfu, was isolated from the hyperthermophilic archaeon Pyrococcus furiosus and introduced in 1991. Pfu exhibits an error rate of approximately 1.3 × 10^{-6} mutations per base pair per cycle, representing a 6- to 100-fold improvement in fidelity over Taq DNA polymerase, depending on the assay and conditions. This error rate can be expressed as roughly 1 mutation per 770,000 base pairs per cycle, calculated from phenotypic assays monitoring detectable mutations in amplified lacI gene sequences. Advanced variants like Phusion DNA polymerase, a fusion of a Pfu-like proofreading domain with a double-stranded DNA-binding domain (such as Sso7d), enhance processivity and extension speed while maintaining high fidelity, with error rates over 50-fold lower than Taq. To facilitate TA cloning, which requires 3' A-overhangs, Pfu or Phusion products—typically blunt-ended—are often blended with Taq polymerase or treated post-amplification to add these overhangs without substantially compromising overall fidelity. Protocols for high-fidelity PCR incorporate optimized conditions to maximize accuracy and yield, including magnesium ion concentrations of 1-2 mM to balance and efficiency, and extension times of about 1 minute per kilobase at 72°C to accommodate the slower processivity of enzymes. Additives like betaine (1-2 M) are commonly included to alleviate secondary structures in GC-rich templates, improving amplification uniformity without affecting . These methods were refined alongside Pfu's introduction, emphasizing fewer cycles (typically 25-30) to further reduce cumulative errors. High-fidelity PCR finds primary applications in , where accurate inserts prevent propagation of errors; , enabling precise base substitutions; and next-generation sequencing (NGS) library preparation, where low mutation rates ensure reliable variant calling. For instance, in NGS workflows, the 100- to 1000-fold fidelity advantage over Taq reduces artifactual variants, critical for clinical . While hot-start mechanisms can be combined for enhanced specificity, the core benefit of high-fidelity PCR lies in its activity for error minimization.

Hot-start Enzymes

Hot-start enzymes refer to modified DNA polymerases designed to remain inactive at ambient temperatures during reaction assembly, activating only upon initial heating to approximately 95°C to minimize non-specific primer annealing and extension. This approach enhances PCR specificity by preventing polymerase activity that could lead to primer dimers or off-target products before the denaturation step. Activation occurs through thermal reversal of the inhibition mechanism, restoring full enzymatic function without altering standard PCR cycling parameters. Several types of hot-start formulations exist, including antibody-blocked Taq polymerases, where monoclonal antibodies bind to the enzyme's active site, inhibiting polymerization until the antibodies denature at high temperatures; chemically modified variants, such as those with reversible alterations to amino acid residues (e.g., SureStart Taq from Agilent), which block activity via covalent linkages that cleave at 95°C; and aptamer-inhibited enzymes, utilizing single-stranded DNA or RNA aptamers that reversibly bind the polymerase, dissociating during the initial heating phase. The antibody method, introduced in a seminal 1994 study, demonstrated prevention of pre-PCR mispriming and primer dimerization, particularly beneficial for amplifying low-copy-number targets. These evolved from earlier 1990s chemical hot-start techniques and manual methods, finding key applications in multiplex PCR setups and analyses involving limited template DNA, where non-specific artifacts can otherwise dominate. In terms of performance, hot-start enzymes substantially reduce non-specific amplification—often by more than 90% in challenging reactions—while preserving complete activity post-activation and enabling higher yields of target amplicons. They are also compatible with polymerase blends, supporting hot-start variants for enhanced in applications requiring accurate replication. Compared to manual hot-start approaches, such as wax beads that physically separate until melting during the first , enzyme-based methods provide greater convenience through single-tube reactions, minimizing handling errors and contamination risks. Prominent commercial examples include Platinum II Taq Hot-Start DNA Polymerase from , which employs a chemical modification for robust inhibitor tolerance and universal annealing, and KAPA HiFi HotStart from , utilizing antibody-mediated inhibition for high-fidelity amplification in NGS library preparation and other demanding assays.

Specialized Polymerases

Specialized polymerases in PCR refer to thermostable enzymes derived from extremophilic microorganisms, offering unique functional properties beyond the standard from . These enzymes, often isolated from thermophilic and in the 1990s through environmental sampling of hot springs and geothermal sites, enable enhanced performance in challenging amplifications, such as those involving templates or difficult sequence compositions. One prominent example is Tth DNA polymerase from Thermus thermophilus, which exhibits dual activity as both a DNA-dependent and a , allowing RNA-dependent cDNA synthesis at elevated temperatures of 60–70°C. This intrinsic functionality requires Mn²⁺ ions and facilitates one-step RT-PCR protocols where reverse transcription and amplification occur in a single tube, reducing contamination risks and simplifying workflows for detection. Tth polymerase supports amplification of targets up to at least 1 kb, making it valuable for analysis and viral diagnostics. Recent engineered variants, such as I640F/I709K (2023), enable Mn²⁺-independent activity for improved flexibility. Vent DNA polymerase, sourced from the archaeon Thermococcus litoralis, provides moderate fidelity with an error rate around 10⁻⁵ mutations per , coupled with exceptional that withstands temperatures exceeding 95°C. Its robust activity on GC-rich or structured templates stems from the enzyme's archaeal origins, enabling reliable of complex genomic regions that challenge standard polymerases. Similarly, Deep Vent DNA polymerase from Pyrococcus sp. GB-D shares these archaeal traits, excelling in high-GC content amplifications due to its high processivity and stability in specialized buffers, often yielding products up to 6 kb with improved specificity over bacterial enzymes. KOD DNA polymerase from Thermococcus kodakaraensis KOD1 stands out for its high processivity, extending up to 100–130 nucleotides per second—several times faster than Taq—allowing ultra-long PCR of fragments exceeding 20 kb with high accuracy (error rates of 10⁻⁶ to 10⁻⁷). This property arises from the enzyme's optimized structure, often combined with exonuclease-deficient mutants for even longer extensions in genomic applications like large inserts. Optimization for these polymerases typically involves adjustments, such as Tricine-based formulations for Tth to enhance RT activity or manganese-free conditions for fidelity in Vent and KOD, ensuring maximal yield and minimal artifacts. Recent examples include the 2024 Neq2X7 polymerase, capable of amplifying long and GC-rich DNA with high fidelity while tolerating dUTP substitution. In the 2020s, engineered variants of these specialized polymerases have been developed for integration with CRISPR-Cas systems, enhancing point-of-care diagnostics by combining isothermal amplification with Cas-mediated detection for rapid identification. These modifications, often involving chimeric fusions or activity-tuned mutants from thermophilic sources, improve in resource-limited settings without thermal cycling.

Isothermal and Alternative Mechanism Variants

Loop-mediated Isothermal Amplification (LAMP)

(LAMP) is a amplification technique that enables rapid and specific amplification under constant temperature conditions, eliminating the need for thermal cycling equipment. Developed in 2000 by Notomi and colleagues, LAMP utilizes a strand-displacing to continuously extend primers and form loop structures on the target DNA, resulting in exponential amplification without denaturation steps. This method has become widely adopted for point-of-care diagnostics due to its simplicity and robustness in resource-limited settings. The LAMP reaction typically employs four to six primers that recognize six to eight distinct regions on the target DNA sequence. The core primers include two outer primers (F3 and B3) and two inner primers (forward inner primer, FIP; backward inner primer, BIP), with optional loop primers (forward loop, LF; backward loop, LB) added to accelerate the process by hybridizing to newly formed stem-loop structures. The reaction relies on the Bst DNA polymerase large fragment, which possesses strong strand displacement activity, and is performed isothermally at 60–65°C for 30–60 minutes, yielding up to 10^9 copies of the target sequence. This efficiency surpasses traditional PCR in speed while maintaining comparable sensitivity, detecting as few as 10 copies of template DNA. Detection of LAMP products can be achieved through multiple methods, including real-time monitoring of turbidity caused by magnesium pyrophosphate precipitation as a byproduct of the reaction, fluorescent dyes such as , or colorimetric indicators like hydroxynaphthol blue for visual readout. Lateral flow assays enable simple, equipment-free detection similar to tests. LAMP has been extensively applied in field diagnostics, such as for caused by , where it provides rapid results in endemic areas with minimal infrastructure. Key advantages of include its isothermal nature, requiring only a heat block or water bath, and high specificity derived from multiple primer binding sites, which reduces off-target amplification compared to methods like (RPA) that rely on recombinase-mediated strand invasion. A variant, reverse transcription (RT-LAMP), incorporates to amplify targets in a single tube, facilitating detection of RNA viruses such as dengue or SARS-CoV-2. However, limitations include the complexity of primer design due to the need for multiple specifically positioned , which can lead to non-specific amplification if not optimized; this issue is often mitigated by adjusting dNTP concentrations and reaction ratios. As of 2025, recent advancements in LAMP include improved visualization techniques, such as enhanced fluorescence and turbidity-based readouts for faster, more sensitive point-of-care applications in viral diagnostics.

Recombinase Polymerase Amplification (RPA)

Recombinase polymerase amplification (RPA) is an isothermal nucleic acid amplification technique that enables rapid, specific detection of DNA targets without the need for thermal cycling, operating at a constant temperature to facilitate point-of-care diagnostics. Developed in 2006 by Piepenburg et al., RPA achieves exponential amplification through a recombinase-mediated strand invasion mechanism, where primers invade double-stranded DNA templates, followed by polymerase extension to displace and amplify new strands. This process mimics homologous recombination, allowing amplification in as little as 10-20 minutes with high sensitivity, detecting as few as 1-10 copies of target DNA. The core components of RPA include the T4 bacteriophage UvsX recombinase, which forms a nucleoprotein filament with primers to enable homologous pairing and strand invasion; single-stranded (SSB) from , which stabilizes displaced strands; and the Bsu polymerase from , a strand-displacing enzyme that extends primers without requiring activity. Reactions typically proceed at 37-42°C, making RPA suitable for resource-limited settings as it avoids specialized equipment like thermocyclers. Unlike (LAMP), which relies on multiple primers to form stem-loop structures, RPA uses a simple two-primer system for efficient invasion and displacement. RPA has been widely applied in point-of-care testing, including rapid detection of pathogens such as virus during the 2015 outbreak in , where field-deployable assays confirmed cases in under 20 minutes. It also supports paper-based diagnostic formats, integrating amplification and readout on low-cost substrates for portable, inhibitor-tolerant tests in remote environments. Detection methods include fluorescence monitoring via probes or end-point lateral flow strips for visual readout, with multiplexing capabilities up to four targets using spectrally distinct probes or barcoded primers. Recent enhancements in the 2020s have combined RPA with CRISPR-Cas systems, such as in the platform, where RPA pre-amplifies targets before Cas13a-mediated collateral cleavage for enhanced specificity and single-molecule detection in complex samples. These hybrids improve discrimination of single nucleotide polymorphisms and enable colorimetric or lateral flow readouts, expanding RPA's utility in infectious and . As of 2025, RPA continues to advance in point-of-care applications for , with new master mixes enabling ultra-rapid detection under ambient conditions.

Helicase-dependent Amplification (HDA)

Helicase-dependent amplification (HDA) is an isothermal amplification technique that employs a DNA helicase enzyme to unwind double-stranded DNA (dsDNA), thereby generating single-stranded templates for primer annealing and extension by a DNA polymerase, all without the need for thermal cycling. This method mimics the replication process by using ATP-dependent activity to separate DNA strands at constant temperatures, typically between 37°C and 65°C, making it suitable for simple, portable devices. Single-stranded binding proteins (SSBs), such as T4 gene 32 protein, stabilize the unwound strands and prevent reannealing, facilitating continuous amplification cycles. HDA was first developed in 2004 by Vincent et al., who demonstrated its use with Escherichia coli UvrD helicase and exo⁻ Klenow fragment polymerase, achieving over 10⁶-fold amplification of target DNA in 3 hours at 37°C. Subsequent optimizations incorporated bacteriophage T7 gene 4 helicase (gp4), which enables unwinding at higher temperatures and supports strand-displacement synthesis with T7 DNA polymerase. This system can yield up to 10⁸-fold amplification within 90 minutes, depending on target length and reaction conditions, with real-time monitoring showing a doubling time of approximately 3 minutes. A key advantage is its ability to amplify from crude samples, such as bacterial cells or blood, without prior DNA purification. Applications of HDA include rapid detection in portable diagnostics, where its isothermal nature eliminates the need for expensive thermocyclers, making it ideal for low-resource settings. It has been integrated into microfluidic microchips for on-site testing of genetic markers, such as in and infectious disease screening, with detection limits comparable to . For RNA targets, reverse transcription HDA (RT-HDA) combines with the system in a one-step reaction, enabling sensitive of viral RNA, as shown in assays for tomato spotted wilt virus and SARS-CoV-2. Detection methods encompass real-time fluorescence using intercalating dyes or labeled primers, as well as endpoint , with the former allowing quantitative monitoring in under an hour. A notable variation is thermophilic HDA (tHDA), which uses thermostable T7 gp4 and Bst to operate at 65°C, enhancing reaction speed and specificity for longer targets up to 2.5 while reducing non-specific amplification. This version improves efficiency in integrated devices and has been applied in multiplex formats for simultaneous detection of multiple . Overall, HDA's simplicity and compatibility with miniaturization position it as a versatile tool for point-of-care . As of 2025, advancements include engineered that further reduce amplification times and integrate HDA into unified biosensing platforms for streamlined detection.

References

  1. [1]
    Full article: PCR Past, Present and Future - Taylor & Francis Online
    Here we review the history of PCR development and the technologies that have evolved from the original PCR method.
  2. [2]
    [PDF] DIFFERENT TYPES OF PCR TECHNIQUES AND ITS APPLICATIONS
    Review Article. ABSTRACT. Polymerase chain reaction is a biological technology to produce ample number of DNA copies of a particular sequence. Three primary ...
  3. [3]
    The History of PCR | Thermo Fisher Scientific - US
    Hot-start techniques were introduced in the late 1980s as a solution to overcome performance issues associated with Taq DNA polymerase. Methods of hot-start PCR ...Missing: review | Show results with:review
  4. [4]
    Hot Start PCR
    ### Summary of Activation Methods for Hot-Start PCR
  5. [5]
    Hot Start PCR - an overview | ScienceDirect Topics
    Hot-start PCR is specifically designed to keep the enzyme inactive until the temperature of the reaction is at least higher than the melting temperature of ...
  6. [6]
    Hot start PCR - PubMed
    Five different Hot Start activation protocols are presented. The first method presents the use of barriers as a means of segregating key reaction components.
  7. [7]
    Using aptamers to control enzyme activity: Hot Start Taq and beyond | NEB
    ### Summary of Aptamer-Based Hot-Start: Advantages and Activation Temperature
  8. [8]
    Comparing Hot Start vs. Standard PCR - AAT Bioquest
    These methods use physical, solid, barriers like wax pellets to segregate reaction components until the hot start activation temperature is achieved. At ...
  9. [9]
    'Touchdown' PCR to circumvent spurious priming during gene ...
    'Touchdown' PCR to circumvent spurious priming during gene amplification. Nucleic Acids Res. 1991 Jul 25;19(14):4008. doi: 10.1093/nar/19.14.4008.
  10. [10]
    'Touchdown' PCR to circumvent spurious priming during gene ...
    R.H. Don, P. T. Cox, B.J. Wainwright, K. Baker, J. S. Mattick; 'Touchdown' PCR to circumvent spurious priming during gene amplification, Nucleic Acids Rese.
  11. [11]
    How is "Touchdown PCR" used to increase PCR specificity? - QIAGEN
    In subsequent cycles, the annealing temperature is decreased in steps of 1–2°C/cycle until a temperature is reached that is equal to, or 2–5°C below, the Tm of ...
  12. [12]
    Touchdown PCR: Easy Intro Plus 5 Expert Tips - Bitesize Bio
    Touchdown PCR is a technique to limit non-specific amplification by starting with a high annealing temperature and slowly decreasing this over PCR cycles.
  13. [13]
    Multiplex-Touchdown PCR to Simultaneously Detect ...
    This study aimed to develop a multiplex-touchdown PCR method to simultaneously detect 3 species of protozoan parasites, ie, Cryptosporidium parvum, Giardia ...
  14. [14]
    An optimized eDNA protocol for fish tracking in estuarine environments
    Jan 7, 2025 · Environmental DNA (eDNA) is revolutionizing how we investigate biodiversity in aquatic and terrestrial environments.
  15. [15]
  16. [16]
    Polymerase Chain Reaction: Basic Protocol Plus Troubleshooting ...
    May 22, 2012 · The development of the polymerase chain reaction (PCR) is one of ... Methods in Enzymology. Vol. 218. San Diego: Academic Press, Inc ...Missing: variants review
  17. [17]
    Nested PCR - Creative Biolabs
    Nested PCR was first used by Saiki et al. in 1988 to detect the human immunodeficiency virus (HIV) and later became widely used in genetic disease diagnosis ...Missing: Simmonds | Show results with:Simmonds
  18. [18]
    Detection of serum hepatitis B virus using a nested polymerase ...
    ... PCR provides a > 10(4) fold increase over the slot-blot assay. Most of the HBe Ag-positive patients were positive in PCR, and most of those being anti-HBe ...
  19. [19]
    An advanced method of asymmetric PCR and its uses in quantitative ...
    Asymmetric PCR potentially circumvents the problem of amplicon strand reannealing by using unequal primer concentrations (5). Depletion of the limiting primer ...
  20. [20]
    Linear-After-The-Exponential (LATE)-PCR: Primer design criteria for ...
    Traditional asymmetric PCR uses conventional PCR primers at unequal concentrations to generate single-stranded DNA. This method, however, is difficult to ...Abstract · Materials And Methods · Results
  21. [21]
    Simple and Effective Method for Generating Single-Stranded DNA ...
    A simple and efficient PCR method was developedfor generating dye- or radiolabeled single-stranded DNA targets or probes used for hybridization studies.Asymmetric Pcr For... · Asymmetric Pcr For Northern... · Microarray Spotting Of Dna...
  22. [22]
    Multiplex Asymmetric PCR-Based Oligonucleotide Microarray for ...
    Traditional asymmetric PCR uses conventional PCR primers at unequal concentrations to generate ssDNA. Unfortunately, this method is often inefficient and ...Materials And Methods · Primers And Probes · Discussion
  23. [23]
    Kinetic PCR analysis: real-time monitoring of DNA amplification ...
    We describe a simple, quantitative assay for any amplifiable DNA sequence that uses a video camera to monitor multiple polymerase chain reactions (PCRs) ...Missing: development | Show results with:development
  24. [24]
    Kinetic PCR Analysis: Real-time Monitoring of DNA Amplification ...
    Sep 1, 1993 · We describe a simple, quantitative assay for any amplifiable DNA sequence that uses a video camera to monitor multiple polymerase chain reactions (PCRs) ...
  25. [25]
    Real-time PCR based on SYBR-Green I fluorescence
    Oct 13, 2003 · We have designed a new assay based on SYBR-Green I binding that is quick, reliable, easily optimised and compares well with the published assay.
  26. [26]
    3' exonuclease activity of Thermus aquaticus DNA polymerase
    The 5'----3' exonuclease activity of the thermostable enzyme Thermus aquaticus DNA polymerase may be employed in a polymerase chain reaction product detection ...
  27. [27]
    [PDF] Real-Time PCR Applications Guide - Bio-Rad
    The cycle number at which this occurs is called the threshold cycle, or CT. Since the CT value is measured in the exponential phase when reagents are not ...
  28. [28]
    PCR efficiency | Determining amplification efficiencies
    ... efficiency (E) for each target gene in a PCR experiment. This formula is expressed as: E = 10^(–1/S) – 1 (where S = slope of the standard curve). To compare ...
  29. [29]
    Analysis of relative gene expression data using real-time ... - PubMed
    The 2(-Delta Delta C(T)) method is a convenient way to analyze the relative changes in gene expression from real-time quantitative PCR experiments.
  30. [30]
    Analysis of Relative Gene Expression Data Using Real-Time ...
    The 2 −ΔΔC T method is a convenient way to analyze the relative changes in gene expression from real-time quantitative PCR experiments.
  31. [31]
    Digital PCR: A brief history - PMC - NIH
    Digital PCR for quantification of a target of interest has been independently developed several times, being described in 1990 and 1991 using the term “limiting ...Missing: seminal | Show results with:seminal
  32. [32]
    Evaluation of a Droplet Digital Polymerase Chain Reaction Format ...
    Nov 28, 2011 · Droplet digital polymerase chain reaction (ddPCR) is a new technology that was recently commercialized to enable the precise quantification ...
  33. [33]
    High Sensitivity Detection and Quantitation of DNA Copy Number ...
    Jan 31, 2014 · For example, digital PCR can be used to detect mutations, to analyze copy number variations seen in the amplifications or deletions of ...
  34. [34]
    Applications of Digital PCR for Clinical Microbiology - PMC - NIH
    May 23, 2017 · Compared to qPCR, dPCR provided more accurate quantitation of pathogens in the presence of inhibitors in the samples, including detection of ...
  35. [35]
    Digital PCR and Real-Time PCR (qPCR) Choices for Different ...
    Robust quantification (high tolerance to PCR inhibitors), Economical running costs. Rapid, simple optimization, Rapid time to results. Not reliant on standard ...
  36. [36]
    [PDF] Overview and recommendations for the application of digital PCR
    Jul 17, 2018 · Digital PCR possesses a number of advantages compared to conventional endpoint PCR and qPCR: • The major advantage of the dPCR method is ...
  37. [37]
    Replacing PCR with COLD-PCR enriches variant DNA sequences ...
    Apr 13, 2008 · Therefore, replacing PCR with COLD-PCR that increases the mutation detection limits does not entail substantial additional investment. The large ...
  38. [38]
    COLD-PCR: improving the sensitivity of molecular diagnostics assays
    COLD-PCR is a PCR-based method that enriches minority alleles by using a lower denaturation temperature, improving sensitivity of molecular diagnostics.Missing: Milano | Show results with:Milano
  39. [39]
    Application of COLD-PCR for improved detection of KRAS mutations ...
    COLD-PCR successfully detected mutations in all samples that were positive by conventional PCR, and enhanced the mutant-to-wild-type ratio by >4.74-fold, ...
  40. [40]
    Ice-COLD-PCR enables rapid amplification and robust enrichment ...
    This simple principle enables COLD-PCR to amplify mutation-containing alleles with a several-fold selectivity over WT alleles (9). Advantages of COLD-PCR ...
  41. [41]
    PCR: celebrating 40 years of development - BioTechniques
    Jan 19, 2023 · Reverse transcriptase was discovered in 1970 by Howard Temin (University of Wisconsin–Madison, WI, USA) and David Baltimore (Salk Institute in ...The Original Pcr · Quantitative Pcr (qpcr) · Digital Pcr (dpcr)
  42. [42]
    M-MuLV reverse transcriptase: Selected properties and improved ...
    Nov 22, 2021 · The presence of RNAse H activity decreases by 4 °C optimal temperature of the M-MuLV RT polymerase activity. H– M-MuLV RT also demonstrates the ...
  43. [43]
    [PDF] M-MLV Reverse Transcriptase, RNase H Minus, Point Mutant ...
    Incubation at elevated temperatures (i.e., 55°C) can decrease RNA secondary structure and enhance cDNA synthesis of some templates. 2. The M-MLV RT Reaction ...
  44. [44]
    One-Step vs Two-Step RT-PCR | Thermo Fisher Scientific - US
    One-step RT-PCR combines RT and PCR in one tube, while two-step RT-PCR performs them separately in different tubes.Missing: procedure | Show results with:procedure
  45. [45]
    COVID-19 Diagnosis: A Comprehensive Review of the RT-qPCR ...
    This review is to shed light on the natural history, pathology, molecular biology, and efficient diagnostic methods of COVID-19, detecting SARS-CoV-2 in ...
  46. [46]
    Analytical Sensitivity and Specificity of Two RT-qPCR Protocols for ...
    In the limit of detection experiment, the N1 and RdRP (modified) showed the highest analytical sensitivity for their RNA targets, 21 and 33.7 copies/reaction, ...<|separator|>
  47. [47]
    RNasin® Ribonuclease Inhibitor - Promega Corporation
    Use RNasin® Ribonuclease Inhibitors for RNA isolation, RT-PCR, in vitro transcription or any situation where RNase contamination is a concern. We offer ...Missing: challenges | Show results with:challenges
  48. [48]
    Optimizing methodologies for PCR-based DNA methylation analysis
    The primary limitation of this technique is that it is qualitative (Citation11). In general, well-standardized MSP assays provide information restricted to ...<|control11|><|separator|>
  49. [49]
    MGMT Gene Silencing and Benefit from Temozolomide in ...
    Mar 10, 2005 · Patients with glioblastoma containing a methylated MGMT promoter benefited from temozolomide, whereas those who did not have a methylated MGMT promoter did not ...
  50. [50]
    real‐time PCR assay for DNA‐methylation using ... - Oxford Academic
    This paper introduces a novel real‐time methylation assay called HeavyMethyl. ... ) Methylation‐specific PCR: a novel PCR assay for methylation status of CpG ...Missing: original | Show results with:original
  51. [51]
    Pyrosequencing versus methylation-specific PCR for assessment of ...
    Jul 31, 2019 · In summary, both PYR and MSP are reliable methods for detecting MGMT methylation in tumor tissue and can be useful for identifying patients ...
  52. [52]
    A modified method of ligation-mediated genome walking for plant ...
    Here we describe a reliable and efficient genome walking method that is particularly effective for plants with large genomes. Our ligation-mediated PCR method, ...
  53. [53]
    Ligation‐Mediated PCR for Genomic Sequencing and Footprinting
    Apr 1, 1991 · This unit describes a ligation-mediated single-sided polymerase chain reaction (PCR) ... Mueller and Wold (1989). Ligase dilution solution, per ...
  54. [54]
    Cyclic Digestion and Ligation-Mediated PCR Used for Flanking ...
    Feb 26, 2020 · Ligation-mediated PCR includes three major steps: (i) restriction enzyme digestion of the genomic DNA and ligation of specific adapters; (ii) ...
  55. [55]
    Efficient chromosomal transposition of a Tc1/mariner - PNAS
    Flanking sequences at the novel integration sites of transposon were amplified by means of ligation-mediated PCR (or splinkerette-PCR) as described (10, 15) and ...
  56. [56]
    High-definition mapping of retroviral integration sites identifies active ...
    We used ligation-mediated polymerase chain reaction (LM-PCR) and pyrosequencing to build a genomewide, high-definition map of > 32 000 MLV integration sites ...
  57. [57]
    Relative quantification of 40 nucleic acid sequences by multiplex ...
    We describe a new method for relative quantification of 40 different DNA sequences in an easy to perform reaction requiring only 20 ng of human DNA.
  58. [58]
    LUMI-PCR: an Illumina platform ligation-mediated PCR protocol for ...
    Feb 4, 2020 · This design reduces the number of PCR cycles required and improves relative quantitation of integration abundance for saturating sequencing coverage.
  59. [59]
    Analysis of any point mutation in DNA. The amplification refractory ...
    Abstract. We have improved the “polymerase chain reaction” (PCR) to permit rapid analysis of any known mutation in genomic DNA. We demonstrate a system, AR.
  60. [60]
    Analysis of any point mutation in DNA. The amplification refractory ...
    Apr 11, 1989 · We have improved the "polymerase chain reaction" (PCR) to permit rapid analysis of any known mutation in genomic DNA.
  61. [61]
    ARMSprimer3: An open-source primer design Python program for ...
    Apr 15, 2025 · In addition to the allele-specific mismatch at the 3′ end, ARMS-PCR introduces additional deliberate mismatches near the 3′ end of the allele- ...
  62. [62]
    A Novel Allele-Specific PCR Protocol for the Detection of the HLA-C ...
    Feb 9, 2021 · A two-step AS-PCR protocol, using four primer sets, was designed to specifically amplify and differentiate the HLA-C*03:02 allele from 17 other ...
  63. [63]
    PCR conditions | Primer annealing specificity | PCR buffers - QIAGEN
    In PCR, annealing occurs between the primers and complementary DNA sequences in the template. Primer annealing must be specific for successful amplification.
  64. [64]
    Touchdown PCR for increased specificity and sensitivity ... - PubMed
    Touchdown (TD) PCR offers a simple and rapid means to optimize PCRs, increasing specificity, sensitivity and yield, without the need for lengthy optimizations.
  65. [65]
    Allele-Specific PCR for KRAS Mutation Detection Using Phosphoryl ...
    ... allele is lower than that of the perfect match allele. The advantages of this method are low cost, high sensitivity (~1% of allele frequency is detectable) ...
  66. [66]
    Validation of a Multiplex Allele-Specific Polymerase Chain Reaction ...
    The MAS-PCR assay detected mutant alleles down to 1% for the G12S mutant ... In this study, we successfully developed highly sensitive and specific MAS-PCR ...<|control11|><|separator|>
  67. [67]
    [PDF] Comparison of allele-specific PCR, created restriction-site PCR, and ...
    The aim of this study was to compare sensitivity, specificity, positive and negative predictive values, accuracy, and rapidity of allele-specific polymerase ...
  68. [68]
    Allele-Specific Quantitative PCR for Accurate, Rapid, and Cost ...
    We developed a one-step open-source genotyping method based on quantitative PCR. The allele-specific qPCR (ASQ) does not require post-PCR processing.Missing: pharmacogenomics | Show results with:pharmacogenomics
  69. [69]
    Multiplex Polymerase Chain Reaction - ScienceDirect.com
    Since the first technique description by Chamberlain in 1988 (Chamberlain et al., 1988) many systems have been developed, including assays to amplify ...
  70. [70]
    Designing highly multiplex PCR primer sets with Simulated ... - Nature
    Apr 11, 2022 · We present and experimentally validate Simulated Annealing Design using Dimer Likelihood Estimation (SADDLE), a stochastic algorithm for design of multiplex ...
  71. [71]
  72. [72]
    Rapid PCR of STR markers: Applications to human identification
    Multiplex PCR with fluorescently labeled primers has been an essential method for the amplification of short tandem repeats used in human identify testing.Missing: panels | Show results with:panels
  73. [73]
    Overcoming the Challenges of Multiplex PCR - Biocompare
    Oct 23, 2012 · Multiplex PCR challenges include complex primer design, competition between targets, and the need for different amplicon sizes or dyes, making ...
  74. [74]
    Multiplex touchdown PCR assay to enhance specificity and ...
    Apr 19, 2019 · The aim of this study was to develop a multiplex touchdown PCR (multiplex TD-PCR) for rapid and simultaneous detection of four major ...
  75. [75]
    [PDF] Multiplex polymerase chain reaction: a practical approach. - SciSpace
    The development of an efficient multiplex PCR usually requires stra- tegic planning and multiple attempts to op- timize reaction conditions. For a successful.<|control11|><|separator|>
  76. [76]
    PCR inhibition in qPCR, dPCR and MPS—mechanisms and solutions
    Feb 12, 2020 · This review includes mechanisms of specific PCR inhibitors as well as solutions to the inhibition problem in relation to cutting-edge DNA analysis.Pcr Inhibition In Qpcr, Dpcr... · Inhibition Of Dna... · Dna Polymerase Inhibitors
  77. [77]
    Inverse Polymerase Chain Reaction - Springer Nature Experiments
    The inverse polymerase chain reaction (IPCR) was the first extension of the conventional polymerase chain reaction to allow the amplification of unknown ...Missing: original paper
  78. [78]
    Inverse Polymerase Chain Reaction | Nature Biotechnology
    Aug 1, 1990 · Ochman, H., Medhora, M., Garza, D. and Hartl, D.L. 1989. In: PCR: Application & Protocols. M. Innis, D. Gelfand, J. Sninsky, ...
  79. [79]
    Mapping Transposon Insertion Sites by Inverse Polymerase Chain ...
    May 2, 2024 · Here we describe how inverse PCR can be used to identify the site of transposon insertion in the bacterial chromosome.Missing: viral | Show results with:viral
  80. [80]
    DNA sonication inverse PCR for genome scale analysis of ...
    Sep 22, 2020 · Inverse PCR (Ochman et al., 1988) is a variant of PCR that has historically been used to obtain flanking sequences (Nowrouzi et al., 2006 ...1 Introduction · 3 Results · 4 Discussion
  81. [81]
    An Inverse PCR Technique to Rapidly Isolate the Flanking DNA of ...
    Aug 6, 2025 · Restriction enzyme mediated integration is a widely used and effective method for insertional mutagenesis in Dictyostelium discoideum.
  82. [82]
    Inverse Polymerase Chain Reaction - an overview
    IPCR (Inverse PCR) is a basic genome walking method based on conventional PCR. It was developed by Ochman et al. (1988), and is the earliest genome walking ...
  83. [83]
    5′-Long Terminal Repeat-Selective CpG Methylation of Latent ...
    To enable the differential analysis of the 5′ LTR, we determined the 5′ flanking DNA sequences of the integrated provirus by using the inverse PCR technique (40) ...
  84. [84]
    High-fidelity amplification using a thermostable DNA polymerase ...
    These studies demonstrate that the error rate per nucleotide induced in the 182 known detectable sites of the lacI gene was 1.6 × 10−6 for Pfu DNA polymerase, ...
  85. [85]
    PCR fidelity of pfu DNA polymerase and other thermostable DNA ...
    Average error rates (mutation frequency/bp/duplication) increased as follows: Pfu (1.3 x 10(-6)) < Deep Vent (2.7 x 10(-6)) < Vent (2.8 x 10(-6)) < Taq (8.0 x ...
  86. [86]
  87. [87]
    Betaine improves the PCR amplification of GC-rich DNA sequences
    It is suggested that betaine improves the amplification of these genes by reducing the formation of secondary structure caused by GC-rich regions.Missing: additive | Show results with:additive
  88. [88]
    [PDF] High-fidelity amplification using a thermostable DNA polymerase ...
    The average error rate was determined to be 1.6 x 10deh for Pfu DNA polymerase and 2.0 x 10 I' for 7'aq DNA polymerase. Thus, if a 1-kb sequence is amplified ...
  89. [89]
  90. [90]
    Error Rate Comparison during Polymerase Chain Reaction by DNA ...
    We have measured the error rates for 6 DNA polymerases commonly used in PCR applications, including 3 polymerases typically used for cloning applications ...
  91. [91]
    How is Hot-Start Technology Beneficial For Your PCR
    Hot-start modifications inhibit DNA polymerase's activity at room temperature, preventing spurious bands from nonspecific amplification.Missing: noise fold
  92. [92]
    PCR Using Hot Start Taq DNA Polymerase | アジレント
    ### SureStart Taq DNA Polymerase Mechanism and Benefits
  93. [93]
  94. [94]
    PCR Amplification | An Introduction to PCR Methods
    The polymerase chain reaction (PCR) is a relatively simple technique that amplifies a DNA template to produce specific DNA fragments in vitro.
  95. [95]
    Platinum II Taq Hot-Start DNA Polymerase - Thermo Fisher Scientific
    Discover Platinum II Taq Hot-Start DNA Polymerases for PCR applications such as genotyping, gene expression profiling, and molecular diagnostics.
  96. [96]
    KAPA HiFi HotStart ReadyMix - Roche Sequencing Store
    KAPA HiFi HotStart ReadyMix is a novel, single-enzyme system that exhibits industry-leading fidelity and performance vs other polymerases.
  97. [97]
    Overview of thermostable DNA polymerases for classical PCR ...
    During the genomics era, the use of thermostable DNA polymerases increased greatly. Many were identified and described-mainly of the genera Thermus, ...
  98. [98]
    Hot Spring Metagenomics - MDPI
    Apr 25, 2013 · The classical example is Taq DNA polymerase from Thermus aquaticus, purified and isolated from hot springs [1], which made the development of ...
  99. [99]
    Manganese-independent Reverse Transcriptase Activity of Tth DNA ...
    The DNA polymerase of Thermus thermophilus (Tth pol) presents reverse transcriptase activity with Mn2+, and can be used for one-step RT-qPCR. However, Mn2+ ...
  100. [100]
    Tth DNA Polymerase - CustomBiotech from Roche
    Tth DNA Polymerase is a thermostable DNA Polymerase with intrinsic reverse transcriptase activity for RT-PCR amplification of RNA to a length of at least 1 kb.
  101. [101]
    rTth DNA Polymerase - TOYOBO
    Therefore, this enzyme enables "one-step RT-PCR" including the reverse transcription and PCR steps. Kits for one-step RT-PCR (Code No. PCR-311F) and real-time ...
  102. [102]
  103. [103]
    Fidelity of Thermococcus litoralis DNA polymerase (Vent) in PCR ...
    DNA synthesis fidelities of two thermostable DNA polymerases, Thermus aquaticus (Taq) and Thermococcus litoralis (Tli, also known as Vent), ...
  104. [104]
  105. [105]
    Advantages of thermococcus kodakaraenis (KOD) DNA Polymerase ...
    Processivity, the number of nucleotides extended before the polymerase detaches, is 5–20 times higher for KOD compared to Taq or Pfu. This property along with ...Missing: scotoductus | Show results with:scotoductus
  106. [106]
    Thermophilic Nucleic Acid Polymerases and Their Application in ...
    Nov 29, 2022 · PCR technique based on KOD DNAP was further developed for accurate amplification of long DNAs [88]. With a mixture of wild-type KOD DNAP and its ...
  107. [107]
    Expression and Characterization of the RKOD DNA Polymerase in ...
    Thermococcus kodakaraensis KOD1 is a DNA polymerase that is widely used in PCR. The DNA coding sequence of KOD was optimized based on the codon usage bias of P.
  108. [108]
    DNA polymerases in precise and predictable CRISPR/Cas9 ...
    Dec 8, 2023 · We found that Polλ is the main DNA polymerase responsible for fill-in of the 5′ overhangs of staggered Cas9 cleavage ends.
  109. [109]
    CRISPR/Cas12a-mediated Enzymatic recombinase amplification for ...
    May 15, 2023 · Combining the CRISPR/Cas12a system with isothermal amplification can overcome the limitation imposed by the poor specificity of isothermal ...
  110. [110]
    Loop-mediated isothermal amplification of DNA - PMC - NIH
    Advantages of LAMP. (i) LAMP amplifies DNA with high efficiency under isothermal conditions without a significant influence of the co-presence of non-target DNA ...Missing: limitations | Show results with:limitations
  111. [111]
    Accelerated reaction by loop-mediated isothermal amplification ...
    Loop-mediated isothermal amplification (LAMP) is a novel nucleic acid amplification method that amplifies DNA with high specificity, efficiency and rapidity ...Missing: paper | Show results with:paper
  112. [112]
    Loop‐mediated isothermal amplification (LAMP) - PubMed Central
    Loop‐mediated isothermal amplification (LAMP) amplifies DNA with high specificity, efficiency and rapidity under isothermal conditions by using a DNA polymerase ...
  113. [113]
    Detection of Loop-Mediated Isothermal Amplification Reaction by ...
    A large amount of by-product, pyrophosphate ion, is produced, yielding white precipitate of magnesium pyrophosphate in the reaction mixture.
  114. [114]
    Evaluation of Loop-Mediated Isothermal Amplification (LAMP) for ...
    These results indicate that LAMP is an effective tool for malaria diagnosis at a field clinic in a field setting. A rapid and accurate diagnosis of malaria ...
  115. [115]
    Real-Time Reverse Transcription Loop-Mediated Isothermal ...
    The present study describes the development of a one-step, single-tube real-time accelerated RT-LAMP assay for rapid detection of the E gene of WN virus.
  116. [116]
    Evolution of the Probe-Based Loop-Mediated Isothermal ... - NIH
    Apr 24, 2023 · The major limitation is that LAMP frequently yields spurious amplicons if the primers interact with each other [35], which can result in false ...
  117. [117]
    Loop-mediated isothermal amplification establishment for detection ...
    The LAMP reaction conditions were optimized by varying the concentration of MgSO4, dNTPs, primers, amplification temperature and reaction time. The results ...Missing: limitations complexity non-
  118. [118]
    DNA Detection Using Recombination Proteins | PLOS Biology
    The technology presented in this study, recombinase polymerase amplification (RPA), overcomes the technical difficulties posed by current DNA amplification ...
  119. [119]
    Recombinase polymerase amplification: Basics, applications ... - PMC
    Recombinase polymerase amplification (RPA) is a highly sensitive, isothermal technique operating at 37-42°C, amplifying as low as 1-10 DNA copies in under 20 ...
  120. [120]
    Recombinase Polymerase Amplification (RPA) - US
    It is performed at a constant temperature of 39–42°C, making it highly suitable for detecting various pathogens in field conditions. RPA can also be used to ...
  121. [121]
    Development and deployment of a rapid recombinase polymerase ...
    In the absence of a vaccine or specific treatments for Ebola virus disease (EVD), early identification of cases is crucial for the control of EVD epidemics.
  122. [122]
    A Paper and Plastic Device for Performing Recombinase ...
    RPA is an isothermal nucleic acid amplification method that utilizes recombinase enzyme to facilitate the binding of oligonucleotide primers to template DNA.
  123. [123]
    Multiplex Detection of Nucleic Acids Using Recombinase ...
    Here, we investigate expanding RPA POC detection by identifying a generic multiplex RPA format that can be combined with a generic multiplex lateral flow device ...
  124. [124]
    SHERLOCK: Nucleic acid detection with CRISPR nucleases - NIH
    Apr 1, 2020 · Here we provide step-by-step instructions for setting up SHERLOCK assays with recombinase-mediated polymerase pre-amplification of DNA or RNA, ...
  125. [125]
    Nucleic acid detection with CRISPR-Cas13a/C2c2 | Science
    Apr 13, 2017 · Of the methods explored, recombinase polymerase amplification (RPA) (18) afforded the greatest sensitivity and could be coupled with T7 ...
  126. [126]
  127. [127]
  128. [128]