Fact-checked by Grok 2 weeks ago

Selected reaction monitoring

Selected reaction monitoring (SRM), also known as multiple reaction monitoring (MRM), is a targeted technique employed in to quantitatively detect and measure predefined peptides or proteins within complex biological samples. It operates on triple instruments, where the first selects a specific precursor by (m/z), the second serves as a collision cell to fragment the , and the third filters and detects specific product , forming unique precursor-product transitions for high selectivity and sensitivity. This method excels in quantifying low-abundance analytes, achieving detection limits as low as 10-50 attomoles on-column, with a linear spanning five orders of magnitude, making it ideal for analyzing proteins in or where concentrations vary widely. SRM's advantages include superior across samples, reduced from complex matrices through dual mass filtering, and the ability to multiplex hundreds to thousands of transitions in a single run when scheduled by retention time. Quantitative accuracy is enhanced by using isotope-labeled internal standards, such as heavy peptides, for absolute or relative measurements. Key applications of SRM span biomarker discovery and validation, particularly in clinical settings like cardiovascular diseases and cancer, where it verifies candidate proteins from with high confidence. It is also used for studying post-translational modifications, , and multi-disease screening assays, offering a cost-effective alternative to antibody-based methods with better capabilities. Recent advances, including software tools like for assay development, have expanded SRM's throughput and applicability in large-scale targeted .

Background and Principles

Definition and Overview

Selected reaction monitoring (SRM), also known as multiple reaction monitoring (MRM), is a targeted tandem mass spectrometry (MS/MS) technique designed for the sensitive and specific quantification of predefined analytes, such as peptides or small molecules, in complex biological samples. In SRM, a specific precursor ion is isolated in the first mass analyzer based on its mass-to-charge ratio (m/z), subjected to fragmentation, and then a specific product ion is selectively monitored in the second mass analyzer to generate a quantifiable signal corresponding to the target analyte. Key elements of SRM include the precursor ion, which represents the intact molecular ion of interest selected by its precise m/z value; collision-induced dissociation (CID), the process by which the precursor ion collides with inert gas molecules in a collision cell to produce fragment ions; and the product ion, a characteristic fragment whose m/z is monitored to confirm identity and enable quantification through ion transitions (pairs of precursor and product m/z values). The basic workflow involves introducing the sample via liquid chromatography, ionizing analytes to form precursor ions, isolating and fragmenting them, and detecting the resulting product ions in a rapid, repetitive cycle to build chromatographic peak intensities for analysis, all without scanning across the full mass range. Unlike discovery-based approaches such as data-dependent acquisition (DDA), which stochastically select abundant ions for fragmentation and are prone to variability in complex mixtures, SRM is hypothesis-driven and focuses exclusively on user-defined transitions, providing higher , selectivity, and by filtering out interfering signals. This targeted nature allows SRM to detect analytes at attomole levels (e.g., 10-50 amol on-column) even in highly complex samples like lysates or , where low-abundance targets might otherwise be obscured. SRM is widely applied in for absolute quantification of proteins via proteotypic peptides.

Historical Development

The origins of selected reaction monitoring (SRM) trace back to the late , when advancements in enabled targeted ion analysis. In 1978, R. Graham Cooks and colleagues, including Richard W. Kondrat and Gary A. McCluskey, coined the term "selected reaction monitoring" and demonstrated its utility for direct analysis of mixtures by selecting and detecting specific fragment ions from precursor ions in tandem MS experiments. Around the same time, in 1978, Richard A. Yost and Christie G. Enke at developed the triple quadrupole mass spectrometer (published in 1979), which allowed for sequential mass selection and fragmentation, forming the instrumental basis for monitoring specific precursor-to-product ion transitions. This innovation built on earlier concepts of selective ion monitoring but extended it to reaction pathways, enhancing specificity in complex mixtures. During the , SRM gained prominence in pharmaceutical analysis, particularly for quantifying drug metabolites and , where its high sensitivity and selectivity proved advantageous over single-stage methods. Commercialization of triple quadrupole instruments by companies like Finnigan (now Thermo Fisher) facilitated broader adoption, shifting SRM from research novelty to routine quantitative tool for small molecules in biological matrices. By the , refinements in triple quadrupole technology, including improved ion optics and software for transition optimization, expanded SRM's applicability to more challenging samples, marking an early foray into through targeted quantification in protein digests. A pivotal in SRM's evolution for occurred in 2004, when Leigh Anderson and colleagues developed the SISCAPA (Stable Isotope Standards and Capture by Anti-Peptide Antibodies) method, combining SRM with immunoaffinity enrichment to quantify low-abundance proteins, enabling detection of over 100 proteins in human with attomolar sensitivity. This work highlighted SRM's potential for large-scale, reproducible protein measurement, catalyzing its integration into biomarker discovery workflows. In the mid-2000s, SRM transitioned to regulated clinical applications, with FDA clearances such as the 2007 approval of liquid chromatography-tandem MS kits for of immunosuppressants in transplant patients. Post-2010, SRM evolved further through hybridization with high-resolution platforms, such as -Orbitrap systems, which improved mass accuracy and multiplexing capacity while retaining targeted quantification; for instance, parallel reaction monitoring (PRM) emerged in 2011 as a high-resolution analog to SRM, allowing simultaneous monitoring of all fragment ions without triple limitations.

Technical Aspects

Instrumentation Requirements

Selected reaction monitoring (SRM) experiments require a (QqQ MS) as the primary instrument, consisting of three quadrupole analyzers in series: the first quadrupole () selects the precursor , the second (q2) serves as a collision cell for fragmentation typically via (), and the third (Q3) selects and detects the specific product . This configuration enables the targeted monitoring of predefined precursor-to-product transitions with high selectivity by filtering out interfering s at both mass selection stages. Key operational parameters for QqQ instruments in SRM mode include unit mass resolution (approximately 0.7 Thomson full width at half maximum, Th FWHM) in Q1 and Q3 to isolate ions differing by at least 0.5-1 Da, distinguishing SRM from full-scan modes that offer broader mass analysis but lower specificity. Dwell time per transition, the duration the instrument spends acquiring data for each ion pair, is typically set between 10 and 50 ms to balance sensitivity and throughput, allowing cycle times of 1-3 seconds for monitoring 50-200 transitions while ensuring at least 8-10 data points across chromatographic peaks of 20-30 seconds width. Auxiliary components are essential, including a liquid chromatography (LC) frontend—often reversed-phase nano-LC with flow rates of 200-500 nL/min—for separating complex peptide mixtures prior to ionization, and an electrospray ionization (ESI) source, preferably nano-ESI, for efficient sample introduction into the mass spectrometer under atmospheric pressure. Instrument control and data acquisition rely on vendor-specific software, such as Thermo Scientific's Xcalibur for TSQ series instruments or Waters' MassLynx for Xevo and Quattro systems, which facilitate method setup, transition scheduling, and real-time optimization of parameters like collision energy. Minimum performance specifications for reliable SRM quantification include a linear dynamic range of 4-5 orders of magnitude (e.g., from femtomolar to micromolar concentrations), high linearity (R² > 0.99 over the range), and limits of detection (LOD) in the low attomolar range (10-50 amol on-column), which are achieved through precise instrument tuning of ion optics, source voltages, and gas pressures.

Experimental Workflow

The experimental workflow for selected reaction monitoring (SRM) in commences with , where complex protein mixtures are enzymatically digested, typically using , to produce peptides that are unique to target proteins (proteotypic peptides). This step avoids peptides prone to missed cleavages or chemical modifications, such as those containing or residues, to ensure stability and consistent ionization. For detecting low-abundance analytes, enrichment methods like immunoaffinity purification or are applied to deplete high-abundance proteins and concentrate targets, enhancing detection limits in biological matrices such as . Assay development follows, involving the selection of precursor-to-product ion transitions derived from prior MS/MS spectral libraries or discovery proteomics data. Tools such as software facilitate this process by importing spectral libraries, predicting transitions (usually 3–5 per for redundancy), and optimizing parameters like fragment selection based on intensity and specificity to minimize interferences. Scheduled acquisition windows are often incorporated to focus monitoring on predicted retention times, enabling higher throughput for multiplexed assays targeting dozens to hundreds of . Samples are then analyzed via liquid chromatography-mass spectrometry (LC-MS), with nanoLC gradients typically spanning 5–60 minutes (e.g., 5–40% acetonitrile) at flow rates of 200–500 nL/min to resolve peptides with sufficient peak capacity and sensitivity. Acquisition parameters on triple quadrupole instruments include monitoring 20–100 transitions per cycle, with dwell times of 10–50 ms per transition to capture chromatographic peaks adequately, and collision energies tuned linearly to precursor m/z values (e.g., CE = 0.044 × m/z + 5.5 eV for doubly charged peptides, typically ranging 10–50 eV). During the LC-MS run, quality checks involve real-time assessment of signal stability through consistent transition intensities and verification of chromatography alignment via co-elution of multiple transitions per , ensuring at least 8–10 data points across each for accurate . Deviations, such as drifting retention times or noisy baselines, prompt immediate adjustments or run termination to maintain data reliability.

Applications

Proteomics

In bottom-up proteomics, selected reaction monitoring (SRM) is employed to monitor specific precursor-to-product ion transitions of proteotypic peptides, which serve as reliable proxies for quantifying the abundance of their parent proteins in complex biological samples. These proteotypic peptides are unique sequences that uniquely identify a protein, minimizing interference from isoforms or related proteins. Typically, 1–3 such peptides are selected per target protein to ensure robust verification and accurate inference of protein levels, as this approach balances specificity, sensitivity, and coverage while accounting for potential peptide-level variability in ionization efficiency or enzymatic digestion. A key aspect of SRM workflows in involves the use of synthetic stable isotope-labeled (SIL) peptides as internal standards for absolute quantification. The absolute quantification (AQUA) strategy, for instance, incorporates heavy isotope-labeled versions of target peptides (e.g., with ^{13}C or ^{15}N) spiked into the sample prior to digestion and analysis; these co-elute and co-fragment with endogenous light peptides, allowing precise determination of protein copy numbers via the ratio of light-to-heavy signals. This method enhances accuracy in dynamic range-limited samples like , where protein concentrations span over 10 orders of magnitude. SRM plays a pivotal role in and validation within , particularly for developing multiplexed panels to detect disease-associated proteins in clinical samples. For example, SRM assays have been optimized to quantify (PSA) and related peptides in diagnostics, enabling differentiation between benign and malignant conditions with high specificity in or extracts. These targeted panels facilitate verification of candidate s identified from , transitioning them toward clinical utility. Notable case studies demonstrate SRM's capability for large-scale proteome mapping, such as quantifying low-abundance proteins in human plasma, revealing insights into the plasma proteome's complexity. Further advancements include integration with selected ion monitoring (SIM) in hybrid workflows to boost sensitivity for trace-level peptides by pre-filtering precursors before fragmentation, as shown in studies achieving reproducible detection in depleted plasma fractions. Scheduled SRM further supports high throughput, allowing simultaneous monitoring of hundreds of transitions across an LC-MS run—up to 100–500 targets—via time-scheduled acquisition windows that optimize dwell times and reduce cycle constraints for multiplexed analysis.

Metabolomics and Other Areas

In metabolomics, selected reaction monitoring (SRM) enables the targeted quantification of small molecule metabolites, such as amino acids and lipids, facilitating pathway analysis in biological samples. For instance, SRM has been applied to monitor tricarboxylic acid (TCA) cycle intermediates like citrate and α-ketoglutarate in disease states, providing insights into metabolic dysregulation. Methods using hydrophilic interaction liquid chromatography (HILIC) coupled with SRM can profile up to 200 polar metabolites involved in central carbon pathways, enhancing the understanding of flux alterations. High-resolution SRM variants have quantified 55 serum metabolites, including branched-chain amino acids, with limits of detection in the nanomolar range for clinical metabolomic studies. Pharmaceutical applications of SRM focus on studies, particularly assays for (CYP450) enzymes that metabolize small molecules under 1000 Da. SRM-based liquid chromatography-tandem (LC-MS/MS) methods simultaneously quantify metabolites from probe substrates for nine CYP450 isoforms, such as acetaminophen from for , supporting high-throughput inhibition screening in drug-drug interaction assessments. In pharmacokinetic (PK) profiling, SRM facilitates precise measurement of parent drugs and metabolites, as demonstrated in validated assays for modulators like and tezacaftor in human plasma, achieving linearity over three orders of magnitude. Beyond metabolomics and pharmaceuticals, SRM extends to food safety, environmental monitoring, and clinical toxicology. In food safety, SRM detects pesticide residues, such as organophosphates in vegetables, using targeted LC-MS/MS transitions for over 400 compounds with minimal matrix interference. For environmental analysis, SRM quantifies polycyclic aromatic hydrocarbons (PAHs) like benzopyrene in airborne particulates and sediments via electron ionization-MS/MS in multiple reaction monitoring mode, aiding pollution tracking. In clinical toxicology, SRM supports therapeutic drug monitoring (TDM) of antibiotics, such as β-lactams in plasma, enabling dose optimization to prevent toxicity while ensuring efficacy. Adaptations of SRM for non-polar compounds include atmospheric pressure chemical ionization (APCI), which improves ionization efficiency for lipophilic small molecules compared to electrospray ionization. Transition lists in SRM workflows typically cover 50-200 small molecules, as in comprehensive serum metabolome assays with 1890 transitions for 595 analytes. Regulatory frameworks endorse SRM in LC-MS bioanalysis; the FDA guidance on bioanalytical method validation (revised 2018) recommends it for PK studies, emphasizing selectivity and sensitivity criteria like signal-to-noise ratios exceeding 10:1 for lower limits of quantification. Similar EMA guidelines highlight SRM's role in validating methods for small molecule quantification in regulatory submissions.

Multiple Reaction Monitoring

Multiple reaction monitoring (MRM), often used interchangeably with selected reaction monitoring (SRM) in the context of triple quadrupole mass spectrometry, extends SRM by simultaneously monitoring multiple predefined precursor-product ion transitions within a single liquid chromatography-mass spectrometry (LC-MS) run, enabling the quantification of numerous analytes from complex biological samples. This targeted approach isolates a specific precursor ion in the first quadrupole (Q1), fragments it in the second (q2), and detects the resulting product ion in the third (Q3), with each transition defined by a unique precursor-to-product mass-to-charge (m/z) pair optimized for the analyte of interest. Technical implementation of MRM requires careful management of acquisition parameters to ensure sufficient sampling across chromatographic peaks. The cycle time, which is the total duration to acquire all transitions in a full scan cycle, must be shorter than the chromatographic peak width—typically calculated as the product of the number of transitions and dwell time per transition, aiming for 10–15 data points per peak to maintain quantification accuracy. For instance, with dwell times of 10–100 ms per transition, a cycle time of approximately 2 seconds supports effective monitoring of 100–200 transitions while resolving peaks of 20–30 seconds full width at half maximum. To accommodate larger numbers of transitions without compromising sensitivity, scheduled MRM confines acquisition to retention time windows around expected elution times for each analyte, allowing up to 1,000 transitions in a single analysis by dynamically adjusting the monitoring schedule based on prior chromatographic data. Optimization of MRM assays focuses on selecting and tuning transitions for maximal specificity and sensitivity. Typically, 3–5 transitions per analyte are chosen, prioritizing the most intense product ions (e.g., y- or b-type fragments) from empirical fragmentation spectra to confirm identity and enhance detection limits, while avoiding interferences. Collision energy is optimized individually or via ramps—scanning a range of energies (e.g., 10–50 eV) during method development—to maximize fragment ion yield, often following empirical formulas like CE = 0.036 × (precursor m/z) + 3.3 for peptides, which can improve signal intensity by 2–5-fold over fixed values. Compared to single-transition SRM, MRM offers substantially higher throughput, enabling the simultaneous analysis of over 100 analytes in one run, which is particularly valuable for multiplexed assays in resource-limited settings. This scalability supports applications like clinical quantification of metabolites, where MRM on triple systems measures 25-hydroxyvitamin D levels with high precision and specificity in , aiding in the of deficiency-related disorders. In newborn screening, MRM facilitates panel-based testing for over 50 metabolic disorders by monitoring multiple transitions for , acylcarnitines, and other biomarkers in dried blood spots, dramatically expanding screening coverage from single-analyte methods.

Parallel Reaction Monitoring

Parallel reaction monitoring (PRM) is a targeted mass spectrometry technique that simultaneously monitors all product ions generated from a selected precursor ion using high-resolution instruments, such as Orbitrap or time-of-flight (TOF) analyzers, enabling the acquisition of complete MS/MS spectra for enhanced specificity in quantification. This approach contrasts with traditional selected reaction monitoring by leveraging high mass accuracy to distinguish interferences without relying on predefined fragment transitions. PRM is typically performed on hybrid quadrupole-Orbitrap systems, like the Q-Exactive, where the isolates the precursor , fragmentation occurs in the collision cell, and the resulting product ions are analyzed at high resolution in the analyzer. These instruments provide mass resolutions exceeding 17,500 and mass accuracies below 5 ppm, allowing for the detection of low-abundance analytes in complex samples. Key benefits of PRM include the generation of full MS/MS spectra in each acquisition cycle, which facilitates robust detection and identification through database searching, and eliminates the need to preselect specific transitions, thereby simplifying development. This results in improved specificity and compared to unit-resolution methods, particularly in matrices with high background noise. In the PRM workflow, targeted MS2 scans are scheduled for 10-20 precursors per liquid chromatography run, with isolation windows of 1-2 Da and rapid cycle times enabled by parallelized detection, resembling data-independent acquisition but restricted to predefined targets for focused quantification. Data extraction involves integrating intensities across all observable fragments, often using tools like for alignment and statistical validation. PRM was introduced in 2012 for applications in , particularly for quantifying phosphopeptides in complex biological matrices, where it demonstrated superior performance in detecting low-stoichiometry modifications with minimal false positives. Subsequent studies have expanded its use to absolute quantification of proteins in and validation of candidates, leveraging its high multiplexing capacity.

Data Processing and Analysis

Quantification Strategies

Selected reaction monitoring (SRM) enables both relative and absolute quantification of target analytes, such as peptides in , by monitoring specific precursor-to-product ion transitions. Relative quantification compares signal intensities between samples, often using label-free approaches or ratios derived from light and heavy isotope-labeled analogs to assess changes in abundance. quantification, in contrast, determines precise molar concentrations by spiking known amounts of internal standards, typically stable isotope-labeled peptides (e.g., AQUA or QconCAT peptides), which co-elute with endogenous targets and correct for losses during and analysis. This strategy achieves coefficients of variation (CVs) below 10% and linearity over 4-5 orders of magnitude, with limits of detection (LOD) in the femtomole range per milligram of sample. Calibration curves are constructed to define the linear dynamic range of SRM assays, typically using 5-6 concentration points of the target analyte spiked with a fixed amount of internal standard, yielding regression coefficients (R²) greater than 0.99. The limit of quantification (LOQ) is established when the signal-to-noise ratio (S/N) exceeds 10, ensuring reproducible measurements with CVs under 20%, while the LOD corresponds to an S/N of at least 3. Peak integration quantifies analyte abundance by calculating the area under the curve (AUC) of chromatographic peaks for selected transitions, with baseline correction algorithms applied to subtract noise and ensure at least 8 data points across the elution profile for Gaussian-shaped peaks. Summed AUCs from multiple transitions enhance accuracy by reducing stochastic noise. Normalization strategies in SRM account for technical variability, either to total protein content using housekeeping peptides or to spike-in controls like isotope-labeled standards. For label-free normalization, the median intensity of unchanged proteins serves as a reference, assuming most analytes remain stable across conditions. Statistical models, such as least-squares , fit calibration curves and estimate analyte concentrations from heavy/light ratios, while methods like mitigate matrix effects by sequentially spiking standards into the sample matrix to extrapolate true concentrations and compensate for suppression or enhancement. These approaches ensure robust quantification, with intra- and inter-laboratory CVs of 10-25% in complex biological matrices.

Validation and Quality Control

Validation of selected reaction monitoring (SRM) assays requires rigorous assessment of , accuracy, and specificity to ensure reliable quantification in complex biological matrices. is typically evaluated through the (), with acceptable limits often set at less than 20% for replicate measurements across the analytical range, reflecting the of peak integration and signal response. Accuracy is determined by comparing measured concentrations to nominal values, aiming for deviations within 15% for standards and samples, which confirms the method's trueness over its of up to five orders of magnitude. Specificity is verified by testing, such as spiking samples with potential interferents or using heavy isotope-labeled standards to detect co-eluting ions that could bias results. Quality control (QC) measures in SRM workflows focus on maintaining run-to-run stability and instrument performance, often incorporating interleaved reference samples analyzed every 10 injections to monitor variability. Retention time alignment is a critical QC metric, with acceptable drift limited to less than 2% across runs to prevent misalignment in scheduled SRM methods, achieved through control charts like XmR or for detecting subtle shifts. These measures use standard peptides or protein mixtures as proxies for biological samples, enabling longitudinal tracking of system suitability and early identification of drifts via change-point . Software tools facilitate SRM validation and QC by automating peak detection, statistical scoring, and data review. Open-source platforms like support method development through spectral library integration and audit logs for documenting validation steps, while mProphet provides semisupervised learning for peak scoring and (FDR) estimation using decoy transitions. Commercial tools such as MultiQuant enable efficient peak review with overlay views of multiple transitions to assess quality and detect interference, incorporating algorithms like MQ4 for consistent integration and regulatory-compliant audit trails. Interference handling in SRM involves checks for co-eluting ions using orthogonal transitions—monitoring multiple fragment ions per precursor to confirm specificity—and statistical validation to estimate FDR, typically targeting rates below 1-5% through decoy-based null distributions in tools like mProphet. This approach filters out false positives by parameterizing scores and ensuring confident identifications, particularly in high-throughput experiments. For clinical SRM assays, regulatory compliance follows guidelines from the Clinical and Laboratory Standards Institute (CLSI) and the U.S. Food and Drug Administration (FDA), emphasizing selectivity testing across at least six individual matrices to demonstrate matrix-independent performance. CLSI C62 outlines verification protocols for LC-MS methods, including precision, accuracy, and interference assessments, while FDA bioanalytical guidance requires full validation of selectivity, specificity, and stability in relevant biological matrices to support diagnostic applications.

Advantages and Limitations

Strengths

Selected reaction monitoring (SRM) offers high , enabling the detection of low-abundance analytes in complex biological samples without the need for sample depletion steps. This technique achieves limits of detection as low as 10–50 amol on column for peptides, corresponding to low ng/mL to pg/mL concentrations in , making it suitable for quantifying rare biomarkers that are otherwise challenging to measure. The dual mass filtering in SRM—selecting specific precursor and fragment ions—provides exceptional specificity, minimizing from matrix components and allowing reliable through multiple co-eluting transitions. SRM demonstrates excellent , with consistent transition intensities across multiple instruments and runs, facilitating cross-laboratory comparisons. Interlaboratory studies have reported coefficients of variation below 20% for the majority of targeted peptides in digests, using standardized protocols on triple-quadrupole mass spectrometers. This consistency arises from the predefined, assay-specific parameters that ensure stable quantification regardless of slight variations in instrumentation. In terms of cost-effectiveness, SRM eliminates the need for custom , relying instead on synthetic proteotypic peptides for , which reduces expenses compared to methods like that require antibody production costing $50,000–$100,000 per target. Furthermore, SRM supports high , quantifying hundreds of targets in a single run, in contrast to the single-plex nature of traditional , enabling scalable analysis at lower per-analyte costs. SRM provides superior quantitative accuracy, with a wide linear spanning 10^4 to 10^5-fold when using stable isotope-labeled internal standards for , outperforming gel-based methods like Western blotting that typically cover only 10^2 to 10^3-fold. This range allows precise absolute and relative quantification of proteins across broad concentration differences in complex mixtures, with accuracy enhanced by the inclusion of heavy standards to account for variations in and detection. The speed of SRM assays supports high-throughput applications, with typical liquid chromatography-mass spectrometry run times of minutes per sample, allowing processing of up to 100 samples per week when combined with efficient targeted workflows. This rapid turnaround enables large-scale screening without compromising sensitivity or specificity.

Challenges and Alternatives

One significant challenge in selected reaction monitoring (SRM) is its susceptibility to ion suppression, particularly in complex biological matrices where co-eluting compounds compete for ionization, leading to reduced signal intensity and quantification inaccuracies. This effect is exacerbated in proteomics samples with high protein content, necessitating careful method optimization to maintain accuracy. Additionally, SRM is inherently limited to monitoring predefined, known target peptides, precluding its use for discovery-based proteomics and restricting applications to hypothesis-driven studies. Transition interference poses another hurdle, as overlapping fragment ions from non-target species can compromise specificity, often requiring extensive empirical optimization of precursor-product ion pairs to minimize false signals. Throughput is constrained by instrument cycle times, which limit the number of concurrent transitions; for instance, more than 100 transitions typically demands reduced dwell times per transition, potentially sacrificing and chromatographic . Furthermore, the unit mass of triple quadrupole (QqQ) instruments used in SRM can lead to false positives from isobaric interferences, especially in low-abundance analyte detection. To address these limitations, mitigation strategies include sample cleanup techniques such as (SPE), which removes matrix interferents and reduces ion suppression effects prior to analysis. Higher-resolution variants like parallel reaction monitoring (PRM) offer improved selectivity by enabling full MS/MS spectra acquisition, though at the cost of slightly lower throughput compared to SRM. Competing techniques provide viable alternatives depending on the application. Data-independent acquisition (DIA) methods enable broader proteome coverage without prior target selection, offering discovery potential alongside quantification in complex samples. SWATH-MS, a DIA variant, serves as a hybrid approach that combines untargeted data collection with targeted extraction, facilitating retrospective analysis and reducing the need for upfront optimization. In clinical settings, immunoassays provide higher specificity for routine biomarker detection, often with simpler workflows and lower costs, though they lack the multiplexing depth of SRM. Looking ahead, integration of , such as models for predicting optimal transitions and automating development, holds promise for overcoming knowledge gaps in target selection and enhancing SRM efficiency in post-2020 workflows.

References

  1. [1]
    Selected reaction monitoring for quantitative proteomics: a tutorial
    Oct 14, 2008 · This tutorial explains the application of SRM for quantitative proteomics, including the selection of proteotypic peptides and the optimization and validation ...
  2. [2]
    An Introduction to Advanced Targeted Acquisition Methods - NIH
    Targeted proteomics via selected reaction monitoring (SRM) or parallel reaction monitoring (PRM) enables fast and sensitive detection of a preselected set ...
  3. [3]
    Selected Reaction Monitoring Mass Spectrometry (SRM-MS) in ...
    In stockThis book introduces the technique of mass spectrometry-based targeted proteomics, and discusses the basics of mass spectrometry, assay development ...
  4. [4]
    Full article: The selected reaction monitoring/multiple reaction ...
    Sep 2, 2014 · Selected reaction monitoring, also known as multiple reaction monitoring, is a powerful targeted mass spectrometry approach for a confident ...
  5. [5]
    Selected reaction monitoring for quantitative proteomics: a tutorial
    Oct 14, 2008 · Selected reaction monitoring (SRM) is a non-scanning technique, generally performed on triple-quadrupole (QQQ) instruments in which ...
  6. [6]
    Empowering Clinical Diagnostics with Mass Spectrometry
    Jan 30, 2020 · This mini-review highlights how MS has created a new paradigm in clinical diagnosis, which is growing in importance for public health.
  7. [7]
    Xcalibur Data Acquisition and Interpretation Software
    Use Thermo Scientific Xcalibur software to control, acquire, and interrogate data from Thermo Scientific LC-MS systems and related instruments.Missing: SRM MassLynx
  8. [8]
  9. [9]
    Selected Reaction Monitoring Mass Spectrometry for Absolute ...
    Aug 17, 2015 · Note: Analyte quantification by LC-QqQ-SRM should be linear over a wide dynamic range (~10,000). At the low end, background signal (random ...Missing: linearity | Show results with:linearity
  10. [10]
    Skyline: open source document editor for proteomics experiments
    The Skyline user interface simplifies the development of mass spectrometer methods and the analysis of data from targeted proteomics experiments performed using ...
  11. [11]
    Development and performance evaluation of an ultralow flow ...
    Jun 11, 2014 · The purpose of this work was to develop a nano LC system operable at a flow rate of 20 nL/min, applicable for routine analysis in proteomics ...
  12. [12]
    High-Throughput Large-Scale Targeted Proteomics Assays for ...
    Targeted proteomics is a mass spectrometry-based protein quantification technique with high sensitivity, accuracy, and reproducibility.
  13. [13]
    MRM/SRM page - UWPR
    Selected reaction monitoring (SRM) is a highly sensitive and selective method for the targeted quantitation of peptides and, by inference, the corresponding ...
  14. [14]
    Absolute quantification of proteins and phosphoproteins from cell ...
    Here we present a strategy for the absolute quantification (termed AQUA) of proteins and their modification states.
  15. [15]
    Absolute quantification of protein and post-translational modification ...
    The AQUA (for 'absolute quantification') method allows sensitive and specific targeted quantification of protein and post-translational modifications in ...
  16. [16]
    Development of selected reaction monitoring‐MS methodology to ...
    Jul 30, 2010 · Development of selected reaction monitoring-MS methodology to measure peptide biomarkers in prostate cancer ... PSA and one peptide from ...
  17. [17]
    Development of selected reaction monitoring-MS methodology to ...
    A multiplexed MS methodology, selected reaction monitoring-MS/MS coupled with stable isotope dilution (SID), was developed and tested in both cells lines and ...
  18. [18]
    Enhanced sensitivity for selected reaction monitoring mass ...
    Here we report enhanced detection sensitivity and reproducibility for SRM-based targeted proteomics by coupling a nanospray ionization multicapillary inlet/dual ...
  19. [19]
    Advancements in Mass Spectrometry-Based Targeted Metabolomics ...
    Dec 16, 2024 · This review summarizes recent advances in targeted metabolomics and lipidomics, emphasizing their applications in clinical research.
  20. [20]
  21. [21]
    Pesticide residues in produce analyzed by Targeted MRM HR “full
    Application of LC-MS/MS with multiple reaction monitoring (MRM) has represented the principal workflow for pesticide residues analyses due to the high ...
  22. [22]
    [PDF] Bioanalytical Method Validation - Guidance for Industry | FDA
    May 24, 2018 · The 2001 guidance for industry on Bioanalytical Method Validationwas ... When using liquid chromatography/mass spectrometry (LC/MS) methods, the.Missing: SRM | Show results with:SRM
  23. [23]
    Multiple Reaction Monitoring-based, Multiplexed, Absolute ... - NIH
    MRM is a targeted approach that requires knowledge of the molecular weight of an analyte and its fragmentation behavior under CID. MRM is capable of highly ...
  24. [24]
    Measurement of vitamin D metabolites by mass spectrometry, an ...
    Dec 5, 2018 · The aim of this review is to provide a comprehensive overview about existing LC-MS/MS methods for the measurement of vitamin D metabolites and their analytical ...
  25. [25]
    How mass spectrometry revolutionized newborn screening - PMC
    Jan 30, 2024 · A groundbreaking multiplex assay capable of screening newborns for more than 30 inherited metabolic conditions with just one test.
  26. [26]
    Parallel Reaction Monitoring: A Targeted Experiment Performed ...
    Dec 2, 2015 · PRM is a targeted method of quantification performed using high-resolution hybrid mass spectrometers such as quadrupole-Orbitrap (q-OT) [4,6,16] ...
  27. [27]
    PRM - UWPR
    A freely-available Windows client application for building Selected Reaction Monitoring (SRM) / Multiple Reaction Monitoring (MRM), Parallel Reaction ...
  28. [28]
    [PDF] Parallel reaction monitoring using quadrupole-Orbitrap mass ... - EPFL
    The experiment was carried out on the latest generation of quadrupole-Orbitrap instrument. (Q-Exactive HF) allowing very short transient times (32 ms at 15 000 ...
  29. [29]
    Parallel Reaction Monitoring for High Resolution and High Mass ...
    Here, we investigate the performance of parallel reaction monitoring (PRM) on a high resolution and accurate mass (HR/AM) quadrupole-Orbitrap mass spectrometer ...
  30. [30]
    Parallel Reaction Monitoring for High Resolution and High Mass ...
    First, PRM spectra would be highly specific because all potential product ions of a peptide, instead of just 3–5 transitions, are available to confirm the ...
  31. [31]
    PRM With an Orbitrap Mass Spec: /home/software/Skyline
    Get hands-on experience setting up an acquisition method to quantify 31 peptides from 19 proteins of interest in murine fibroblasts using Parallel Reaction ...
  32. [32]
    The Parallel Reaction Monitoring-Parallel Accumulation–Serial ...
    Jan 18, 2022 · In this study, a prm-PASEF approach was used for the multiplexed absolute quantitation of proteins in human plasma using isotope-labeled peptide standards.Abstract · Subjects · Results And Discussion
  33. [33]
  34. [34]
  35. [35]
    MSstatsQC: Longitudinal System Suitability Monitoring and Quality ...
    Selected Reaction Monitoring (SRM) is a powerful tool for targeted detection and quantification of peptides in complex matrices. An important objective of ...
  36. [36]
    Automated quality control system for LC-SRM setups - PubMed
    Dec 16, 2013 · We present here a quality control workflow that is based on repeated analysis of a standard sample to allow insight into the stability of the ...Missing: measures run- run drift
  37. [37]
    Start Page: /home/software/Skyline
    ### Summary of Skyline's Role in SRM Method Development and Validation
  38. [38]
    MultiQuant Software - SCIEX
    MultiQuant software is a powerful and an easy-to-use quantitation package, which processes MRM, UV, ADC, and full scan data for quantitation.
  39. [39]
    [PDF] mProphet: A general and flexible data model and algorithm for ...
    To estimate false discovery rates (FDR) depending on the discriminant score. (mProphet score) cutoff we parameterize a null distribution based on the decoy ...
  40. [40]
    Protein Significance Analysis in Selected Reaction Monitoring (SRM ...
    Selected reaction monitoring (SRM) is a mass spectrometry technique that can accurately and reproducibly quantify proteins in complex biological mixtures (1, 2, ...
  41. [41]
    C62 | Liquid Chromatography-Mass Spectrometry Methods - CLSI
    CLSI C62 provides comprehensive guidance for developing and verifying liquid chromatography-mass spectrometry (LC-MS) methods in the clinical laboratory.
  42. [42]
    Selected reaction monitoring for quantitative proteomics: a tutorial | Molecular Systems Biology
    ### Summary of Selected Reaction Monitoring (SRM) in Mass Spectrometry
  43. [43]
    Advancing the sensitivity of selected reaction monitoring-based ...
    Selected reaction monitoring (SRM)—also known as multiple reaction monitoring (MRM)—has emerged as a promising high-throughput targeted protein quantification ...Missing: definition | Show results with:definition
  44. [44]
    Platform for Establishing Interlaboratory Reproducibility of Selected ...
    The aim of this study was to establish a complete workflow for assessing the transferability and reproducibility of selected reaction monitoring (SRM) assays.
  45. [45]
    Antibody-Free PRISM–SRM for Multiplexed Protein Quantification
    In general, SRM assays have several distinct advantages over immunoassays due to their fast and cost-efficient assay development process, a multiplexing ...
  46. [46]
    Applications Of Selected Reaction Monitoring (SRM) - PubMed Central
    We illustrate the approach of stable isotopic dilution (SID)-selected reaction monitoring (SRM)-mass spectrometry (MS) assays for quantification of low ...Missing: femtomole | Show results with:femtomole
  47. [47]
    Ion suppression; A critical review on causes, evaluation, prevention ...
    This review article aims to collect together the latest information on the causes of ion suppression in LC-MS analysis and to consider the efficacy of common ...
  48. [48]
    Detection and Correction of Interference in SRM Analysis - PMC - NIH
    We present here an approach to detect interference by using the expected relative intensity of SRM transitions. We also designed an algorithm to automatically ...Missing: optimization | Show results with:optimization
  49. [49]
    Ultrathroughput Multiple Reaction Monitoring Mass Spectrometry
    Jan 11, 2010 · The UMRM technology transforms existing approaches of multiple reaction monitoring (MRM) by a unique integration with peptide derivatization.Supporting Information · Author Information · References
  50. [50]
    Is High Resolution a Strict Requirement for Mass Spectrometry ... - NIH
    QqQ-MS however also produced nine false-positive results owing to the inherent instrumental mass resolution limits. To discriminate the false-positive results ...
  51. [51]
  52. [52]
    [PDF] Parallel Reaction Monitoring: A Targeted Experiment Performed ...
    Dec 2, 2015 · Abstract: The parallel reaction monitoring (PRM) assay has emerged as an alternative method of targeted quantification.Missing: femtomole | Show results with:femtomole
  53. [53]
    Advances in data‐independent acquisition mass spectrometry ...
    May 29, 2022 · The data-independent acquisition mass spectrometry (DIA-MS) has rapidly evolved as a powerful alternative for highly reproducible proteome profiling.
  54. [54]
    Data‐independent acquisition‐based SWATH‐MS for quantitative ...
    Aug 13, 2018 · This tutorial provides guidelines on how to set up and plan a SWATH‐MS experiment, how to perform the mass spectrometric measurement and how to analyse SWATH‐ ...Finding An Optimally... · Peptide Query Parameter... · Peptide And Protein...<|control11|><|separator|>
  55. [55]
    Monoplex and multiplex immunoassays: approval, advancements ...
    Nov 20, 2021 · Immunoassays are a powerful diagnostic tool and are widely used for the quantification of proteins and biomolecules in medical diagnosis and research.Introduction · Lateral Flow Immunoassay · Multiplex Assays As An...
  56. [56]
    LC-SRM Combined With Machine Learning Enables Fast ... - NIH
    This new workflow, called LC-SRM-ML, combines the strengths of capillary flow LC and SRM acquisition to make it suitable for routine analysis in clinical ...