Open access peer-reviewed chapter

Mass Spectrometry in Clinical Laboratories

Written By

Jadranka Miletić Vukajlović and Tanja Panić-Janković

Submitted: 03 June 2020 Reviewed: 19 February 2021 Published: 08 April 2021

DOI: 10.5772/intechopen.96745

From the Edited Volume

Mass Spectrometry in Life Sciences and Clinical Laboratory

Edited by Goran Mitulović

Chapter metrics overview

680 Chapter Downloads

View Full Metrics

Abstract

The analyses performed in clinical laboratories require a high level of precision, selectivity, and sensitivity. The rising number of therapeutic agents from both the field of small and large molecules and the increasing use of modern screening approaches have brought mass spectrometry into almost every clinical laboratory. The need to screen the patients and to follow the therapy’s success can often be fulfilled only by the highly selective and sensitive targeted approach with mass spectrometry. With improving instrument design and miniaturization of the separation technologies, mass spectrometry is no longer an exotic analytical approach. The use of mass spectrometry is now not restricted to the use in a clinical laboratory, but it is used in operating rooms for instant and on-site helping the surgeons with defining the margin of the tissue to be extracted. In this manuscript, we describe the use of mass spectrometry for selected clinical applications and show the possible way of future applications.

Keywords

  • Clinical laboratory
  • antibiotics
  • newborn screening
  • mass spectrometry

1. Introduction

The use of mass spectrometry in the clinical laboratory has become a standard for analysis of different substances such as antibiotics, for newborn screening, detection of immune-suppressive drugs, or the analysis of therapeutic antibodies used for the treatment of different diseases.

The focus of the use of mass spectrometry in clinical settings is the analysis of clinical samples and monitoring levels of active compounds and their metabolites in patients’ blood and urine samples. The high sensitivity and specificity of the mass spectrometer and the possibility to perform specific detection of target analytes by applying MRM/SRM (multiple reaction monitoring/selected reaction monitoring) enable a targeted and highly specific analytical approach. The methods developed need a separation method in front of the MS and several companies such as Chromsystems (https://www.chromsystems.com/), ThermoFisher Scientific (https://www.thermofisher.com/at/en/home/clinical/diagnostic-testing/clinical-chemistry-drug-toxicology-testing/therapeutic-drug-monitoring.html) or BioRad (www.bio-rad.com), to name just a few, have developed fully verified and certified analytical systems. The interested reader is encouraged to search the internet for additional providers and systems.

Applying chromatography and mass spectrometry has its primary values in relatively fast detection and measuring of multiple analytes in a single sample with high sensitivity and high selectivity. In clinical routine, the key challenge for identifying and analyzing active compounds is having the sensitivity of the analytical system needed and required to detect and quantify low-concentration analytes.

One of the challenges for using the MS in a clinical laboratory was the low ion yield, which significantly hampered the development of clinical applications. However, the development of new analytical systems, especially of new ion inlets and ion funnel designs with the most widely used electrospray ionization (ESI) sources has significantly improved ion focusing and ion transfer, which, finally, resulted in the overall increased sensitivity.

The quality of electrospray is highly dependable on separation conditions, i.e. mobile phase, presence or absence of salts, flow speed, column’s inner diameter, etc. In proteomics, the use of columns with 50 μm or 75 μm ID is state-of-the-art. However, the columns operated at a low flow rate of several hundreds of nanoliters/minute are still rare in clinical analysis although they can provide a significant increase in analysis‘s sensitivity.

However, currently, the use of nanoflow separation still cannot cope with the demand for high sample throughput in clinical applications. Currently, the closest compromise between sensitivity and throughput is the use of the microbore and capillary columns of 300 μm – 500 μm and 1 mm – 2 mm inner diameter.

A new and exciting application of mass spectrometry in the clinical environment is the use of “live-MS” during surgical operations. Further development of this approach will revolutionize the diagnostics and help surgeons in extracting e.g. tumors with higher accuracy and higher yield.

Advertisement

2. Clinical applications

2.1 Analysis of antibiotics

Antibiotics, either cytotoxic or cytostatic to the microorganisms, have been widely used to treat and prevent infectious diseases and allow the body’s natural defenses to eliminate them. They usually have a role to inhibit the synthesis of proteins, deoxyribonucleic acid (DNA), ribonucleic acid (RNA), or other specific actions [1]. Using the energy-dependent transport mechanisms in ribosomal sites, antibiotics target bacterial cell wall by attaching to them, which consequently results in inhibition of protein synthesis and subsequent cell death [2].

With the discovery of penicillin by Sir Alexander Fleming, a new, modern, chapter of innovation and antibiotics development began [3]. Today, there are different classes of antibiotics (Table 1) and they are widely used not only in human medicine but also in veterinary medicine and aquaculture [35]. However, antibiotics inadvertently released into the environment can cause a massive threat to the ecosystems and subsequently to human health. Consequently, they may accumulate in food and, which is much more worrying, antibiotic resistance of human pathogens might develop through the transfer of environmental bacteria genes (ARGs) [36, 37, 38, 39, 40]. Furthermore, sensitive individuals might experience allergic reactions triggered by antibiotic residues in food. Furthermore, the ingestion of sub-therapeutic doses of antibiotics and uncontrolled use of antibiotics may initiate the development of drug-resistant strains of bacteria that initially appeared in hospitals only, where most antibiotics were being used [41].

Different classes of antibioticsReferences
β-lactam[4, 5, 6]
Sulfonamides[7, 8, 9]
Aminoglycosides[10, 11, 12]
Tetracyclines[13, 14]
Chloramphenicol[15, 16]
Macrolides[17, 18, 19]
Glycopeptides[20, 21, 22]
Oxazolidinones[23, 24]
Ansamycins[25, 26]
Quinolones[27, 28]
Streptogramins[29, 30]
Lipopeptides[31, 32]
Antibiotic Resistance[33, 34]

Table 1.

Overview of different classes of antibiotics and some related references describing their mass spectrometry analysis.

In recent years, more and more scientific data and news reporting the misuse and the overuse of antibiotics [42, 43], the environment exposure pathways [44, 45], and the presence of antibiotic-resistant strains [46, 47] became available.

Over the years, numerous analytical methods have been developed and described to determine antibiotic residues in the environment and food. Chromatographic separation and detection of antibiotics and their metabolites using various detectors is the most widely used analytical approach for monitoring and determination.

The electrospray ionization technique has become the technique of choice in many areas of analyzing biologically relevant macromolecules [48]. The soft ionization MS techniques - matrix-assisted laser desorption/ionization (MALDI) and ESI [49, 50] proved to be the best approach for analysis due to the efficient ionization of polar antibiotics. Depending on the ionization mode applied, it has been shown that most antibiotics yield a better signal when positive ionization is used, with the most commonly formed protonated molecular ion [M + H]+. Determination of analytes trace levels in complex biological matrices using the molecular ion generally is not enough selective due to the limited resolution of unit-mass MS instruments. Therefore, these obstacles are overcome by using modern equipment consisting of liquid chromatography (LC) MS instrumentation with tandem MS (MS/MS), which became the technique of choice in quantitative bioanalysis. Tandem MS capacities enhance selectivity and signal-to-noise ratio and provide essential structural information based on which it is possible to identify the structural conformation of analyzed samples. For these reasons, many laboratories use triple quadrupole (Q) MS/MS over ion trap (IT) MS instruments in routine practice for detection and analysis of antibiotics and other drug residues [51, 52, 53, 54, 55]. This advantage is reflected in its quantitative features regarding IT MS with its MSn capabilities, which are highly beneficial for analysis of analyte’s molecular structure and identification.

With technological advancement, instruments providing accurate-mass, high-resolution (HR) time-of-flight (TOF) MS, single TOF-MS, or hybrid instruments combined with a quadrupole (Q-TOF-MS) and the collision cell for MS/MS analysis became available. HR-MS has entered every day’s practice of clinical laboratories as a viable alternative to traditional triple quadrupole mass spectrometer. The versatility of HR-MS (especially hybrid HR-MS) is reflected in increased selectivity by eliminating potential interferences originating from the matrix with remarkably similar mass-to-charge ratio (m/z) as of the measured analytes, but with a different structure.

Unlike IT and Q MS, TOF-MS is a pulsed, and a non-scanning MS. TOF-MS can acquire full spectral data, thus separates and detects ions of various m/z by measuring the time taken for the ions to travel through a field-free region. Therefore, these instruments are mostly combined with a fast LC separation if used for rapid non-targeted screening [56].

Based on all the above-mentioned, LC–MS is an essential factor in the pharmaceutical industry and clinical laboratory due to the possibility of identifying impurities in synthetic products, characterize metabolites, and perform quantitative bioanalysis.

The following section of this chapter provides an overview of examples of mass spectrometry usage in clinical laboratories for detection and characterization of antibiotics in a different sample including pharmaceutical, blood (plasma, serum), environmental water samples (waste, surface, and drinking water) [57, 58, 59, 60, 61, 62], animal and plants and products of animal and plant origin, etc. [63, 64, 65].

Depending on sample matrices such as muscle, liver, kidney, egg, milk, or honey, multiclass methods based on LC–MS or LC–MS/MS are used for the analysis of antibiotics residues [66, 67, 68, 69]. The complexity of the methods depends also on the complexity of the sample preparation. Therefore, screening methods try to avoid complicated sample preparation such as solid-phase extraction (SPE) and the evaporation of the purified extract before the chromatographic separation whereas quantitative methods do not bypass this step [70]. Chico et al. [71] analyzed 39 analytes residues that belong to 5 families of antibiotics with different physicochemical properties which include sulfonamides (SAs), quinolones (Qs), tetracyclines (TCs), macrolides (MCs), and penicillins (PCs). To shorten the analysis time, their method set-up was based on ultra-high-pressure liquid chromatography (UHPLC), like in Yamaguchi et al. [72] and Tian et al. [73], combined with tandem mass spectrometry–MS/MS with ESI. Mass spectrometry parameters were determined and optimized by an infusion of standard solutions to accomplish the highest sensitivity. The singly protonated molecular ion was selected and used as the precursor ion for all compounds [M + H]+, and the cone voltage was adjusted to its maximum signal at the first quadrupole of the mass spectrometer. The success of this method proved to be exceptional and, for that reason, was introduced as a method at the laboratory of Agència de la Salut Pública de Barcelona.

Several analytical methods are currently available to separately detect the fluoroquinolone and sulfonamide classes of antibiotics in manure, surface water, wastewater, and groundwater [74, 75, 76]. Haller et al. [77] focused on liquid–liquid extraction followed by LC–MS analysis of veterinary antibiotics (sulfonamides and trimethoprim), which are most commonly being leaked into the aquatic environment. Based on published LC–MS methods for sulfonamides separation on a reversed-phase chromatographic column, ammonium acetate buffered water and acetonitrile were used as mobile phases. The most successful baseline separation was achieved using the buffered mobile phases at pH 4.6, which enables more stable retention times and better peak shapes for almost all analyzed analytes due to their pKa. Analytes appear to be more hydrophobic and retain better on an RP HPLC column. Haller et al. [77] acquired SIM mass spectra of all samples (antibiotics and of the internal standard) in the full scan mode, using positive and negative electrospray ionization. Single-protonated [M + H]+ or the [M–H], and several (two to three) additional fragments that were generated through the in-source fragmentation, which is typical for single quadrupole mass spectrometer and that yielded the best signal-to-noise (S/N) ratios were selected for confirmation. The advantage of this method is multiple: a very simple extraction process was applied, thus sample preparation is faster, the method does not require tandem mass spectrometry, the method is capable of detecting the investigated pharmaceuticals, to determine the half-lives of antibiotics in manure slurry, and to establish mass balances from antibiotic contents in medicinal feed to quantities.

Renew et al. [74] analyzed groups of antibiotics (fluoroquinolones, sulfonamides, and trimethoprim) simultaneously at sub micrograms per liter concentrations in wastewater effluents using readily available LC–MS techniques. Quantification and identification were performed by applying fluorescence detection and additionally confirmed by tandem LC–MS.

Following Chico et al. SPE followed by LC–MS analysis is utilized by this method. Hirsch et al. [78] and Hartig et al. [79] developed LC–MS techniques for sulfonamides detection. The application of this method allowed preliminary determination of the occurrence of these antibiotics in municipal wastewater treatment plants. Usually, normal phase chromatography (NPC), which implied that the use of a polar stationary phase, was used for the LC separation. However, the NP stationary phases usually show large heterogeneity, which was also observed in this experiment as a consequence of peak tailing and non-linear retention factors with varying analyte concentrations [80]. Different solvents were used to accomplish elution in NPC, from non-polar organic to some variants like the use of isohydric solvents [81, 82]. Some obstacles like lack of retention of highly hydrophilic compounds with ionizable functional groups have been exceeded by ion-exchange chromatography [83] or ion pairing on reversed-phase (RP) columns [84]. However, for those analytes with high hydrophilicity the problem has been overcome using hydrophilic interaction chromatography (HILIC). In contrast to the RP LC, the gradient elution in HILIC starts with a low-polarity, low acquoeus organic solvent and elutes polar analytes by increasing the polar content. In addition, in HILIC, ion pair reagents are not required, and the separation system can be easily coupled to MS, especially in the ESI mode [85].

A large topic opens when it comes to antibiotic treatment, as well as establishing resistance to them. MALDI-TOF [33, 86] technique is an ionization technique that allows the analysis of biomolecules and is used to monitor antibiotic treatment as well as rapid detection of antibiotic resistance. The feasibility of MALDI-TOF MS identification of bacterial colonies from solid media has been evaluated on a wide range of clinically relevant bacterial strains as well as yeast isolates.

MALDI-TOF MS whole-cell extracts identification represents a new method for obtaining a characteristic bacterial fingerprint, which allows for distinction of microorganisms based on different genera, species, and from different strains of the same species. The advantages of using this method are numerous: identification can be achieved in a short time after culture isolation, sensitivity is high, ability to detect microorganisms is not limited to prespecified targets, mass spectra obtained for unknown microorganisms are compared with reference database to achieve the identification. Therefore, MALDI-TOF MS represents a reliable method for rapid bacteria and fungi identification in a clinical setting.

The biggest global challenge due to growth rates of multi-drug-resistant microorganisms, especially in hospital settings, introduces new analytical methods not only for prevention and treatment but also for the detection and determination of antibiotic-resistant species. Numerous MALDI-TOF MS-based methods have been recommended for the rapid detection of antibiotic-resistance in bacterial pathogens isolated from bloodstream infections as well as for detection of antimicrobial-resistance in pathogenic fungi. Methods based on an assessment of β-lactamase activity, biomarkers detection responsible for drug-resistance, and/or non-susceptibility, and the comparison of bacteria proteomic profiles incubated with or without antimicrobial drugs, are the most widely studied [33].

2.2 Newborn screening-amino acid analysis

The newborn screening (NBS) program was developed for early diagnosis of asymptomatic newborns at risk for rare diseases such as inborn errors of metabolism (IEM). However, meanwhile, the screening program includes all newborn independently of the risk. IEM is a serious, degenerative, chronic disease with painful and unpredictable clinical manifestations varying from apparent clinical state or obfuscated with other diseases’ symptoms to differing degrees of mental retardation and physical disability [87]. These diseases often result in disturbed levels of amino acids or acylcarnitines, which are used as diagnostic markers for IEM. Many problems correlated to irregular amino acid metabolism generate abnormal ammonia concentrations, resulting in an increased turnover of amino acids for energy production or an indicator of alterations in urea cycle metabolism [88]. In the 1960s, the first NBS for the most frequent aminoacidopathia, which is phenylketonuria (PKU), (Guthrie & Susi, 1963), was developed using a dried blood spot (DBS) [89]. It was established to detect PKU and enable early treatment and prevent neurodevelopmental problems if untreated. From the newborn screening perspective, time is a vital factor in the disease etiology. Without screening, many disorders cannot be recognized on time and untreated patients can exhibit serious symptoms of the disease and end up in a coma or even face death. Children diagnosed on time and with adequate treatment are functional, have reducing sequelae or at least substantially lessening organ damages, and may live normal life [90].

Thirty years ago, the first report was published using tandem mass spectrometry (TMS) [91] for analyzing multiple acylcarnitines and amino acids on a single blood spot. The following development of TMS had been introduced as combined with fast atom bombardment (FAB) and electrospray ionization (ESI) [92, 93, 94, 95, 96] recently with high-resolution liquid or gas chromatography (LC, GC) respectively [97], and direct analysis in real-time [98] mass spectrometry for newborn screening purposes. According to multiple authors [99, 100, 101, 102, 103], blood spot extracts are analyzed by FIA coupled to triple quadrupole (TQ) TMS. Although TQ instruments possess robustness and sensitivity, these instruments also experience monoisotopic interferences with naturally occurring 13C isotopologues, in-source fragmentation interferences, and low mass resolving power, which leads to difficulties separating isobaric compounds with identical quantifying product ions.

The possibility of multiple disorders detection in a single blood spot shortly after birth increased with new technologies in mass spectrometry. TMS is the most widely used instrument for the detection and analysis of amino acids in the DBS and represents one of the most important advancements in the neonatal screening approach [104, 105]. Figure 1 shows a general scheme of DMB sample preparation for MS analysis.

Figure 1.

Example of DMB sample preparation steps for MS analysis [106].

Analysis of specific amino acids proved to be adequate indicator for the presence of certain disorders in newborns. By measuring fluctuations and disturbances in amino acid metabolism, a diverse group of disorders can be identified and confirmed [107]. Disorders that affect the metabolism of amino acids include PKU, tyrosinemia type I (TYR I), maple syrup urine disease (MSUD), homocystinuria (HCY), argininosuccinic aciduria (ASA), and citrullinemia (CIT) (Table 2). These disorders are autosomal recessive and can be confirmed by analyzing amino acid concentrations in body fluids. Because of more than 500 confirmed disorders detected, the use of TMS for clinical screening in a newborn is the method of choice for a few million newborn screenings worldwide [90, 106, 112, 114, 115, 116].

DisordersMarker(s)MethodReferences
Classic phenylketonuria (PKU)Phe, TyrLC–MS/MS[108]
Tyrosinemia type ISA, TyrLC–MS/MS[109]
Marple syrup urine disease (MSUD)Leu, Ile, ValLC–MS/MS[110]
Homocystinuria (HCY)MetMALDI-TOF MS[111]
Argininosuccinic aciduria (ASA)Asa, CitHPLC–MS/MS[112]
Citrullinemia (type I and II) (CIT)CitESI-MS/MS[113]

Table 2.

Exemplary overview of the parameters used in newborn screening and the technology applied.

Phe (phenylalanine), Tyr (tyrosine), SA (succinylacetone), Leu (leucine), Ile (isoleucine), Val (valine), Met (methionine), Asa (argininosuccinic acid), Cit (citrulline).

When it comes to the operation mode of the instrument, TMS can be operated in different modes such as neutral loss scanning, precursor ion scanning, and multiple reaction monitoring. When neutral loss scanning mode is used, all precursors undergoing the loss of a common, neutral, fragment such as water, ammonia, or a phosphate-group are being detected and can be used for further experiments. The neutral loss method is applied for the detection of amino acids due to the neutral loss of m/z 46, which is being shared by many amino acids during fragmentation [117].

Acylcarnitines are detected by this method, as they produce a characteristic fragment ion of 85 m/z. On the other hand, other amino acids, e.g. arginine, ornithine, and citrulline, split off other fragments during collision-induced dissociation. Due to their basic functional group that fragments easily, the most common loss is a combination of butyl formate and ammonia [105, 116, 117, 118, 119]. Therefore, for the detection of all amino acids, acylcarnitines, and other biological compounds in NBS, it is safer and better to use the multiple-reaction-monitoring mode for data acquisition (MRM).

To detect the compound of interest, this method requires individual mass transition optimization to achieve the highest selectivity and sensitivity for both amino acids and acylcarnitines, and only selected amino acids can be measured quantitatively and selectively [116]. It is crucial to emphasize the difference between “screening” and quantification in TMS analysis of amino acids. For the diagnosis of PKU, determination of phenylalanine/tyrosine (Phe/Tyr) is of higher importance than a precise measurement of only phenylalanine [120, 121].

For transient neonatal hypertyrosinemia, an elevated level of tyrosine is usually detected by TMS. To differentiate, the diagnosis of TYR I can be established by detecting the presence of succinylacetone in serum or urine [122]. It is important to emphasize that the high concentration of tyrosine is not always a companion of TYR I [123]. Allard et al. developed a method for verifying TYR1 by using succinylacetone as a determination marker (SUAC) in DBS [124]. Some data reported that this method is unmistakably sensitive and specific, while other reports pointed out that false-positive results were also obtained [125]. Many screening programs for homocystinuria have combined determination of methionine (Met) as a primary marker, methionine, and phenylalanine ratio (Met/Phe), and the total homocysteine (tHcy) as a second-tier marker in DBS [126, 127].

Bartl et al. incorporated the LC–MS/MS analysis as a potential first step in screening clinically symptomatic high-risk populations for the two types of HCY and severe B-vitamin deficiencies. In several IEMs, increased reactive oxygen species (ROS) causes pathophysiological oxidative damage that, in the case of HCY, excess Hcy directly supports ROS formation in the form of O2, hydrogen radical, or H2O2 [128]. Elevated Hcy concentration is deemed a risk factor for neurodegenerative diseases inducing neurological dysfunction via oxidative stress [129]. Mild to moderate increases in Hcy levels have been associated with both vascular dementia and Alzheimer’s disease (AD) [130, 131] and with a possible increased risk of developing Parkinson’s disease at a later age [132, 133, 134].

Many cases on the diagnosis of PKU [135], MSUD [136], and HCY [126] in newborn blood spots using amino acid analysis by FAB TMS were also reported.

Screening of a large number of disorders was established when Rashed et al. [137] used ESI for analyzing butyl esters of amino acids and acylcarnitines. Consequently, clinical laboratories around the world use this automated sample insertion and data analysis method for a newborn screening procedure to detect and analyze selected amino acids and acylcarnitines [138]. Chace et al. [139] first described the use of TMS for MSUD NBS and recommended the determination of total leucine (Xle) in combination with a total leucine and phenylalanine ratio (Xle/Phe, respectively) for improved detection. In the following studies, recommendations for MSUD detection was based on an elevated Xle or leucine (Leu) [112, 140, 141, 142, 143, 144, 145, 146, 147]. Some studies reported that Val is also required for referral [148, 149, 150] while others did report Val, but without the cut-off value [139, 143, 151]. Other studies also included the Xle/alanine (Ala) ratio [151]. In a long 11-years-long study in the Netherlands, MSUD NBS was measured in almost two million newborns using TMS, and MSUD was confirmed for 4 patients and 118 false-positive referrals. The authors recommended Xle/Phe ratio as a promising additional marker ratio to their MSUD NBS strategy and advised consideration of method implementation in the Dutch NBS program [138].

Although sensitive, the newborn screening does have some limitations, and therefore, particular caution is required to the common symptoms that may indicate a metabolic disorder. Its goal is to prevent morbidity and mortality through the early detection of metabolic disorders. A significant number of these disorders may present in the neonatal period; therefore, the need for a newborn screening technique is rising. Tandem mass spectrometry has emerged rapidly in previous years as a crucial multiplex testing technique for biochemical genetics analysis and newborn screening and the number of possible disorders that may be included for NBS has exponentially increased.

2.3 On-site mass spectrometry in OP-room

The continuous increase in the prevalence of cancer requires continuous innovation of both diagnostics and treatment. One of the crucial steps in cancer therapy is as complete as possible surgical removal of the tumor from the surrounding healthy tissue. This so-called negative margin assessment is of critical importance for complete tumor removal and for achieving tumor remission and improve the overall survival rate of patients.

Surgical on-site decision-making could be enhanced with devices and different methods that give an instant and adequate biochemical information about the multiple biopsies or continuous sampling during surgery. Different MS platforms have shown to be able to provide and substantial impact in surgical decision-making process in different points during clinical workflow. To achieve this goal, surgeons would greatly benefit from using mass spectroscopy during the actual operation is going and having immediate information about the resected tumor. This would significantly increase the rate of successfully and almost completely removed tumors and reduce the risk of tumor recurrence. One of the main requirements, or a minimum requirement, for surgery, is that the selected technique delivers fast and accurate information on unprocessed samples and that the ionization is performed as ambient ionization thus eliminating the need for suction and minimizing the use of other solvents than sterile water. DESI (desorption electrospray ionization) was the first technology to be used for the offline analysis of resected tissue. For DESI, a spray of charged solvents is directed onto the tissue’s surface and secondary droplets containing the analytes are desorbed and sampled by the MS. Based on this approach, Eberlin et al. [152, 153] developed the MasSpec Pen for intraoperative MS analyses and rapid diagnostics of cancer.

One example is the use of the MasSpec Pen (MS Pen) for diagnostics of ovarian cancer [154] published by Sans et al. Ovarian cancer is a highly lethal disease that is very often diagnosed very late and it is the fifth leading cause of deaths among women [155, 156]. Furthermore, as with other cancers, accurate diagnosis is of extreme importance for the selection of the treatment and development of precision medicine approach and personalized medicine and therapy. For ovarian cancer, two therapy scenarios are possible: a) cytoreductive surgery before chemotherapy and b) surgery upon chemotherapy for tumors that cannot be fully resected. The timing for the cytoreductive surgery is of great importance and in both cases, it is very important to differentiate the tumor from the healthy tissue with high precision. Identification of a tumor can also be very difficult in cases where scarring or some other fibrous tissue is present and, sometimes, healthy tissue is removed, which should be avoided. Unlike iKnife, the MS Pen uses a water droplet to extract molecules from the tissue [157] and transfer it to the ion source. The full process is very fast, it needs no derivatization or other kinds of sample preparation and the acting surgeon gets an instant result based on a database search, which can help to properly identify the resected tissue and enable better determination of the resection margin. Sans et al. [154] have described the use of MassSpecPen for rapid diagnosis of ovarian cancer. The authors analyzed tissue samples from the tissue bank or from prospectively collected samples from endometriosis surgeries to establish the database needed. The authors analyzed the presence of small metabolites such as glycerophosphoinositol, glycerophosphoserine, glutathione, and glycerophospholipid. It was found that normal ovarian tissue was characterized by presence of ascorbate and some other small metabolites with a relatively high abundance in comparison to cancer samples.

The iKnife was developed with the same purpose as the MassSpec Pen but it relies on ionizing analytes in the smoke plum that is generated during electro cauterization of the tissue during the surgery [158]. Unlike the MassSpec Pen, iKnife is preferably used to identify lipids in the smoke plum. By comparing the mass spectra of the sample generated during the surgery and the database that was established earlier, the surgeon sees the result instantly on the screen and can make decisions about further procedure. St. John et al. described the use of iKnife for the identification of breast pathology for breast cancer surgery [159]. The aerosol produced by the monopolar hand piece used in surgery was aspirated and analytes therein were ionized in the mass spectrometer’s ion source. Generated data were used to identify the tissue by applying multivariate analysis. The method proved to be able to identify the substances within a very short time range of 1.8 seconds. Here, the spectral differences that arise between the two operational modes of the electrosurgical knife – the “cut” and the “clog” – were combined to create a multivariate statistical model and to allow for using both modes during the surgery.

A further application where the iKnife was applied is ex-vivo use for diagnosis of cervical disease. The specimen obtained by cervical punch biopsy can either bee snap frozen and used for confirmation of the conventional histology analysis or it can be analyzed immediately upon sampling. Tzafetas et al. [158] was showed that the application of this technology enabled identification of lipids that characterize cancer, the normal tissue, and samples affected by HPV.

MALDI mass spectrometry has already proved efficient for analyzing microorganism and for the offline imaging (MSI) tissue analyses. It is the MSI that represent an encouraging tool to support histopathology analyses and the decision-making processes. MALDI MSI captures the entire spectrum of biomolecules, including specific biomarkers, providing enhanced discriminating power over the visual inspection of tissue and placing it as a proper assisting method in diagnosis procedure.

With the progress of ambient mass spectrometry techniques, such DESI, MS became a powerful methodology for characterizing lipids within tumor specimens. The DESI MS analysis can be performed with minimal sample preparation and it provides molecular information from tissue samples rapidly. This qualifies the DESI and MALDI methods as a diagnostic method in the OP room. In addition to tumor classification, defining tumor subtypes, and identifying tumor grade, this method also provides necrotic tumor tissue identification, an indicator of high-grade malignancy, and can help distinguish necrotic tumor tissue from viable tumor regions [159, 160, 161, 162].

Advertisement

3. Conclusion

The use of MS in clinical laboratories worldwide increasing, and, as a result, substantial improvements in assay performance are occurring rapidly in many areas such as toxicology, endocrinology, and biochemical genetics. Numerous types of mass spectrometers are being used for the characterization of small molecules such as drugs of abuse, steroids, amines, amino and organic acids, as well as larger compounds such as proteins and ribosomal RNA.

The development of MS technologies has pushed clinical MS toward the analysis of peptides and proteins for diagnostic examination. However, the quantitative analysis of proteins by MS is still a challenging area of laboratory medicine, which faces many challenges before being fit for a routine application. Also, MS contributes to the quality of the many test results (standardization of assays for steroids, lipids, hemoglobin A1c, etc.), and is used as a standard method in all US states for newborn screening. Furthermore, it is important to address that nearly every institution sends tests to the reference laboratories which frequently perform these tests using MS. With the improved functionality that benefits novel front-end modifications and computational abilities, MS can now be used for nontraditional clinical analyses, including clinical microbiology applications for bacteria differentiation and in surgical operating rooms.

We did not address the role of MALDI imaging technology for application in pathology, but it is one of the fastest-growing application of mass spectrometry in clinical settings and the growth is only impeded by a lack of fast and easy to use software packages for fast identification of analytes others than peptides or lipids.

Advertisement

Conflict of interest

The authors declare no conflict of interest.

References

  1. 1. Levy SB, Marshall B: Antibacterial resistance worldwide: causes, challenges, and responses. Nature Medicine. 2004,10:122-129. DOI: 10.1038/nm1145
  2. 2. Maranan MC, Moreira B, Boyle-Vavra S, et al.: Antimicrobial resistance in staphylococci: Epidemiology, molecular mechanisms, and clinical relevance. Infectious Disease Clinics of North America. 1997;11:813-849. DOI: 10.1016/S0891-5520(05)70392-5
  3. 3. Fleming A. On the antibacterial action of cultures of a penicillium, with special reference to their use in the isolation of B. influenzae. The British journal of experimental pathology. 1929;10:223-236
  4. 4. Petrović M, Hernando MD, Díaz-Cruz MS, Barceló D. Liquid chromatography–tandem mass spectrometry for the analysis of pharmaceutical residues in environmental samples: a review. Journal of Chromatography A. 2005 Mar 4;1067(1-2):1-4
  5. 5. Mastovska K, Lightfield AR. Streamlining methodology for the multiresidue analysis of β-lactam antibiotics in bovine kidney using liquid chromatography–tandem mass spectrometry. Journal of Chromatography A. 2008 Aug 22;1202(2):118-123
  6. 6. Yu J, Liu J, Li Y, Yu J, Zhu W, Liu Y, Shen L. Rapid detection of carbapenemase activity of Enterobacteriaceae isolated from positive blood cultures by MALDI-TOF MS. Annals of clinical microbiology and antimicrobials. 2018 Dec 1;17(1):22
  7. 7. Białk-Bielińska A, Kumirska J, Palavinskas R, Stepnowski P. Optimization of multiple reaction monitoring mode for the trace analysis of veterinary sulfonamides by LC–MS/MS. Talanta. 2009 Dec 15;80(2):947-953
  8. 8. García-Galán MJ, Villagrasa M, Díaz-Cruz MS, Barceló D. LC-QqLIT MS analysis of nine sulfonamides and one of their acetylated metabolites in the Llobregat River basin. Quantitative determination and qualitative evaluation by IDA experiments. Analytical and bioanalytical chemistry. 2010 Jun 1;397(3):1325-1334
  9. 9. Yu W, Li K, Liu Z, Zhang H, Jin X. Novelty aqueous two-phase extraction system based on ionic liquid for determination of sulfonamides in blood coupled with high-performance liquid chromatography. Microchemical Journal. 2018 Jan 1;136:263-269
  10. 10. Oertel R, Neumeister V, Kirch W. Hydrophilic interaction chromatography combined with tandem-mass spectrometry to determine six aminoglycosides in serum. Journal of chromatography A. 2004 Nov 26;1058(1-2):197-201
  11. 11. Kumar P, Rubies A, Companyo R, Centrich F. Hydrophilic interaction chromatography for the analysis of aminoglycosides. Journal of separation science. 2012 Feb;35(4):498-504
  12. 12. Sharma D, Kumar B, Lata M, Joshi B, Venkatesan K, Shukla S, Bisht D. Comparative proteomic analysis of aminoglycosides resistant and susceptible Mycobacterium tuberculosis clinical isolates for exploring potential drug targets. PLoS One. 2015 Oct 5;10(10):e0139414
  13. 13. Meisen I, Wisholzer S, Soltwisch J, Dreisewerd K, Mormann M, Müthing J, Karch H, Friedrich AW. Normal silica gel and reversed phase thin-layer chromatography coupled with UV spectroscopy and IR-MALDI-o-TOF-MS for the detection of tetracycline antibiotics. Analytical and bioanalytical chemistry. 2010 Dec 1;398(7-8):2821-2831
  14. 14. Gavilán RE, Nebot C, Miranda JM, Martín-Gómez Y, Vázquez-Belda B, Franco CM, Cepeda A. Analysis of tetracyclines in medicated feed for food animal production by HPLC-MS/MS. Antibiotics. 2016 Mar;5(1):1
  15. 15. Boyd B, Björk H, Billing J, Shimelis O, Axelsson S, Leonora M, Yilmaz E. Development of an improved method for trace analysis of chloramphenicol using molecularly imprinted polymers. Journal of Chromatography A. 2007 Dec 7;1174(1-2):63-71
  16. 16. Rezende DR, Filho NF, Rocha GL. Simultaneous determination of chloramphenicol and florfenicol in liquid milk, milk powder and bovine muscle by LC–MS/MS. Food Additives & Contaminants: Part A. 2012 Apr 1;29(4):559-570
  17. 17. Göbel A, McArdell CS, Suter MJ, Giger W. Trace determination of macrolide and sulfonamide antimicrobials, a human sulfonamide metabolite, and trimethoprim in wastewater using liquid chromatography coupled to electrospray tandem mass spectrometry. Analytical Chemistry. 2004 Aug 15;76(16):4756-4764
  18. 18. Berrada H, Borrull F, Font G, Moltó JC, Marcé RM. Validation of a confirmatory method for the determination of macrolides in liver and kidney animal tissues in accordance with the European Union regulation 2002/657/EC. Journal of Chromatography A. 2007 Jul 20;1157(1-2):281-288
  19. 19. Wang J. Analysis of macrolide antibiotics, using liquid chromatography-mass spectrometry, in food, biological and environmental matrices. Mass spectrometry reviews. 2009 Jan;28(1):50-92
  20. 20. Lin YS, Tsai PJ, Weng MF, Chen YC. Affinity capture using vancomycin-bound magnetic nanoparticles for the MALDI-MS analysis of bacteria. Analytical chemistry. 2005 Mar 15;77(6):1753-1760
  21. 21. König K, Kobold U, Fink G, Leinenbach A, Dülffer T, Thiele R, Zander J, Vogeser M. Quantification of vancomycin in human serum by LC-MS/MS. Clinical Chemistry and Laboratory Medicine (CCLM). 2013 Sep 1;51(9):1761-1769
  22. 22. Oyaert M, Peersman N, Kieffer D, Deiteren K, Smits A, Allegaert K, Spriet I, Van Eldere J, Verhaegen J, Vermeersch P, Pauwels S. Novel LC–MS/MS method for plasma vancomycin: Comparison with immunoassays and clinical impact. Clinica Chimica Acta. 2015 Feb 20;441:63-70
  23. 23. Ji HY, Lee HW, Chang SG, Lee JJ, Rhee JK, Kim WB, Lee HS. Liquid chromatography-tandem mass spectrometry for the determination of a new oxazolidinone antibiotic DA-7867 in human plasma. Biomedical Chromatography. 2004 Mar;18(2):86-89
  24. 24. Wang Y, Li X, Fu Y, Chen Y, Wang Y, Ye D, Wang C, Hu X, Zhou L, Du J, Shen J. Association of florfenicol residues with the abundance of oxazolidinone resistance genes in livestock manures. Journal of Hazardous Materials. 2020 May 28:123059
  25. 25. Jensen MR, Schoepfer J, Radimerski T, Massey A, Guy CT, Brueggen J, Quadt C, Buckler A, Cozens R, Drysdale MJ, Garcia-Echeverria C. NVP-AUY922: a small molecule HSP90 inhibitor with potent antitumor activity in preclinical breast cancer models. Breast Cancer Research. 2008 Apr 1;10(2):R33
  26. 26. Liu LL, Chen ZF, Liu Y, Tang D, Gao HH, Zhang Q, Gao JM. Molecular networking-based for the target discovery of potent antiproliferative polycyclic macrolactam ansamycins from Streptomyces cacaoi subsp. asoensis. Organic Chemistry Frontiers. 2020;7(24):4008-4018
  27. 27. Jansen LJ, Bolck YJ, Rademaker J, Zuidema T, Berendsen BJ. The analysis of tetracyclines, quinolones, macrolides, lincosamides, pleuromutilins, and sulfonamides in chicken feathers using UHPLC-MS/MS in order to monitor antibiotic use in the poultry sector. Analytical and Bioanalytical Chemistry. 2017 Aug 1;409(21):4927-4941
  28. 28. Zhao Y, Tang M, Liu F, Li H, Wang H, Xu D. Highly Integrated Microfluidic Chip Coupled to Mass Spectrometry for Online Analysis of Residual Quinolones in Milk. Analytical chemistry. 2019 Sep 30;91(21):13418-13426
  29. 29. Vrioni G, Tsiamis C, Oikonomidis G, Theodoridou K, Kapsimali V, Tsakris A. MALDI-TOF mass spectrometry technology for detecting biomarkers of antimicrobial resistance: current achievements and future perspectives. Annals of Translational Medicine. 2018 Jun;6(12)
  30. 30. Civitareale C, Mestria S, Gallo P, Giannetti L, Neri B, Stacchini P, Fiori M. Multi-drug ultraperformance liquid chromatography/tandem mass spectrometry method to quantify antimicrobials in feeding stuffs at carry-over level. Rapid Communications in Mass Spectrometry. 2018 Nov 15;32(21):1831-1842
  31. 31. Armbruster KM, Meredith TC. Enrichment of bacterial lipoproteins and preparation of N-terminal lipopeptides for structural determination by mass spectrometry. JoVE (Journal of Visualized Experiments). 2018 May 21(135):e56842
  32. 32. Liu Y, Teng K, Wang T, Dong E, Zhang M, Tao Y, Zhong J. Antimicrobial Bacillus velezensis HC6: production of three kinds of lipopeptides and biocontrol potential in maize. Journal of Applied Microbiology. 2020 Jan;128(1):242-254
  33. 33. Idelevich EA, Sparbier K, Kostrzewa M, Becker K. Rapid detection of antibiotic resistance by MALDI-TOF mass spectrometry using a novel direct-on-target microdroplet growth assay. Clinical Microbiology and Infection. 2018;24:738-743.DOI: 10.1016/j.cmi.2017.10.016
  34. 34. Angeletti S. Matrix assisted laser desorption time of flight mass spectrometry (MALDI-TOF MS) in clinical microbiology. Journal of microbiological methods. 2017 Jul 1;138:20-29
  35. 35. Mastovska, Katerina. “Multiresidue analysis of antibiotics in food of animal origin using liquid chromatography–mass spectrometry.” Mass spectrometry in food safety. Humana Press, 2011. 267-307. DOI: 10.1007/978-1-61779-136-9_12
  36. 36. Bengtsson-Palme J, Larsson DJ. Antibiotic resistance genes in the environment: prioritizing risks. Nature Reviews Microbiology. 2015;13:396. DOI: 10.1038/nrmicro3399-c1
  37. 37. Du L, Liu W. Occurrence, fate, and ecotoxicity of antibiotics in agro-ecosystems. A review. Agronomy for sustainable development. 2012;32:309-327. DOI: 10.1007/s13593-011-0062-9
  38. 38. Li B, Yang Y, Ma L, Ju F, Guo F, Tiedje JM, Zhang T. Metagenomic and network analysis reveal wide distribution and co-occurrence of environmental antibiotic resistance genes. The ISME journal. 2015;9:2490-2502. DOI: 10.1038/ismej.2015.59
  39. 39. Martínez JL, Coque TM, Baquero F. What is a resistance gene? Ranking risk in resistomes. Nature Reviews Microbiology. 2015;13:116-123. DOI: 10.1038/nrmicro3399
  40. 40. Van Boeckel TP, Brower C, Gilbert M, Grenfell BT, Levin SA, Robinson TP, Teillant A, Laxminarayan R. Global trends in antimicrobial use in food animals. Proceedings of the National Academy of Sciences. 2015;112:5649-5654. DOI: 10.1073/pnas.1503141112
  41. 41. Yang Y, Song W, Lin H, Wang W, Du L, Xing W. Antibiotics and antibiotic resistance genes in global lakes: A review and meta-analysis. Environment international. 2018;116:60-73. DOI: 10.1016/j.envint.2018.04.011
  42. 42. Chang Y, Chusri S, Sangthong R, McNeil E, Hu J, Du W, Li D, Fan X, Zhou H, Chongsuvivatwong V, Tang L. Clinical pattern of antibiotic overuse and misuse in primary healthcare hospitals in the southwest of China. PloS one. 2019;14:e0214779. DOI: 10.1371/journal.pone.0214779
  43. 43. Limmathurotsakul D, Sandoe JA, Barrett DC, Corley M, Hsu LY, Mendelson M, Collignon P, Laxminarayan R, Peacock SJ, Howard P. ‘Antibiotic footprint’ as a communication tool to aid reduction of antibiotic consumption. Journal of Antimicrobial Chemotherapy. 2019 Aug 1;74(8):2122-2127. DOI: 10.1093/jac/dkz185
  44. 44. Ben Y, Fu C, Hu M, Liu L, Wong MH, Zheng C. Human health risk assessment of antibiotic resistance associated with antibiotic residues in the environment: a review. Environmental research. 2019;169:483-493. DOI: 10.1016/j.envres.2018.11.040
  45. 45. Singer AC, Shaw H, Rhodes V, Hart A. Review of antimicrobial resistance in the environment and its relevance to environmental regulators. Frontiers in microbiology. 2016 Nov 1;7:1728. DOI: 10.3389/fmicb.2016.01728
  46. 46. Bergeron S, Boopathy R, Nathaniel R, Corbin A, LaFleur G. Presence of antibiotic resistant bacteria and antibiotic resistance genes in raw source water and treated drinking water. International Biodeterioration & Biodegradation. 2015;102:370-374. DOI: 10.1016/j.ibiod.2015.04.017
  47. 47. Ljubojević D, Puvača N, Pelić M, Todorović D, Pajić M, Milanov D, Velhner M. Epidemiological significance of poultry litter for spreading the antibiotic-resistant strains of Escherichia coli. World’s Poultry Science Journal. 2016;72:485-494. DOI: 10.1017/S004393391600043X
  48. 48. Yi X, Bayen S, Kelly BC, Li X, Zhou Z. Improved detection of multiple environmental antibiotics through an optimized sample extraction strategy in liquid chromatography-mass spectrometry analysis. Analytical and bioanalytical chemistry. 2015;407:9071-9083. DOI: 10.1007/s00216-015-9074-7
  49. 49. Wu C, Kim HK, van Wezel GP, Choi YH. Metabolomics in the natural products field–a gateway to novel antibiotics. Drug Discovery Today: Technologies. 2015;13:11-17. DOI: 10.1016/j.ddtec.2015.01.004
  50. 50. Jamshidi-Aidji M, Morlock GE. Bioprofiling of unknown antibiotics in herbal extracts: Development of a streamlined direct bioautography using Bacillus subtilis linked to mass spectrometry. Journal of Chromatography A. 2015;1420:110-118. DOI: 10.1016/j.chroma.2015.09.061
  51. 51. Moldoveanu S, Scott W, Zhu J. Analysis of small carbohydrates in several bioactive botanicals by gas chromatography with mass spectrometry and liquid chromatography with tandem mass spectrometry. Journal of separation science. 2015;38:3677-3686. DOI: 10.1002/jssc.201500573
  52. 52. Huang Z, Pan XD, Huang BF, Xu JJ, Wang ML, Ren YP. Determination of 15 β-lactam antibiotics in pork muscle by matrix solid-phase dispersion extraction (MSPD) and ultra-high pressure liquid chromatography tandem mass spectrometry. Food control. 2016;66:145-150. DOI: 10.1016/j.foodcont.2016.01.037
  53. 53. Gentili A, Perret D, Marchese S. Liquid chromatography-tandem mass spectrometry for performing confirmatory analysis of veterinary drugs in animal-food products. TrAC Trends in Analytical Chemistry. 2005;24:704-733. DOI: 10.1016/j.trac.2005.02.007
  54. 54. Blasco C, Picó Y, Torres CM. Progress in analysis of residual antibacterials in food. TrAC Trends in Analytical Chemistry. 2007;26:895-913. DOI: 10.1016/j.trac.2007.08.001
  55. 55. Panić-Janković T, Mitulović G. Human chorionic gonadotrophin pharmaceutical formulations of urinary origin display high levels of contaminant proteins–A label-free quantitation proteomics study. Electrophoresis. 2019;40(11):1622-1629. DOI: 10.1002/elps.201900087
  56. 56. Mastovska, K. Recent developments in chromatographic techniques, in Comprehensive Analytical Chemistry. In: Pico, Y, editor. Food Contaminants and Residue Analysis. 1st ed. Oxford: Elsevier; 2008. p. 175-200. DOI: 10.1007/978-1-61779-136-9_12
  57. 57. Samanidou VF, Hapeshi EA, Papadoyannis IN. Rapid and sensitive high-performance liquid chromatographic determination of four cephalosporin antibiotics in pharmaceuticals and body fluids. Journal of Chromatography B. 2003;788:147-158. DOI: 10.1016/S1570-0232(02)01040-1
  58. 58. Samanidou VF, Ioannou AS, Papadoyannis IN. The use of a monolithic column to improve the simultaneous determination of four cephalosporin antibiotics in pharmaceuticals and body fluids by HPLC after solid phase extraction—a comparison with a conventional reversed-phase silica-based column. Journal of Chromatography B. 2004;809:175-182. DOI: 10.1016/j.jchromb.2004.06.019
  59. 59. Kim C, Ryu HD, Chung EG, Kim Y, Lee JK. A review of analytical procedures for the simultaneous determination of medically important veterinary antibiotics in environmental water: Sample preparation, liquid chromatography, and mass spectrometry. Journal of environmental management. 2018;217:629-645. DOI: 10.1016/j.jenvman.2018.04.006
  60. 60. Ding J, Ren N, Chen L, Ding L. On-line coupling of solid-phase extraction to liquid chromatography–tandem mass spectrometry for the determination of macrolide antibiotics in environmental water. Analytica Chimica Acta. 2009;634:215-221. DOI: 10.1016/j.aca.2008.12.022
  61. 61. Kim C, Ryu HD, Chung EG, Kim Y. Determination of 18 veterinary antibiotics in environmental water using high-performance liquid chromatography-q-orbitrap combined with on-line solid-phase extraction. Journal of Chromatography B. 2018;1084:158-165. DOI: 10.1016/j.jchromb.2018.03.038
  62. 62. Gracia-Lor E, Sancho JV, Hernández F. Multi-class determination of around 50 pharmaceuticals, including 26 antibiotics, in environmental and wastewater samples by ultra-high performance liquid chromatography–tandem mass spectrometry. Journal of Chromatography A. 2011;1218:2264-2275. DOI: 10.1016/j.chroma.2011.02.026
  63. 63. Vishnuraj MR, Kandeepan G, Rao KH, Chand S, Kumbhar V. Occurrence, public health hazards and detection methods of antibiotic residues in foods of animal origin: A comprehensive review. Cogent Food & Agriculture. 2016;2:1235458. DOI: 10.1080/23311932.2016.1235458
  64. 64. Wegener HC. Antibiotics in animal feed and their role in resistance development. Current opinion in microbiology. 2003;6:439-445. DOI: 10.1016/j.mib.2003.09.009
  65. 65. Gousia P, Economou V, Bozidis P, Papadopoulou C. Vancomycin-resistance phenotypes, vancomycin-resistance genes, and resistance to antibiotics of enterococci isolated from food of animal origin. Foodborne Pathogens and Disease. 2015;12:214-220. DOI: 10.1089/fpd.2014.1832
  66. 66. Rosén J. Efficient and sensitive screening and confirmation of residues of selected polyether ionophore antibiotics in liver and eggs by liquid chromatography-electrospray tandem mass spectrometry. Analyst. 2001;126:1990-1995
  67. 67. Matabudul DK, Conway B, Lumley I, Sumar S. The simultaneous determination of the ionophore antibiotics in animal tissues and eggs by tandem electrospray LC–MS–MS. Food Chemistry. 2001;75:345-354. DOI: 10.1016/S0308-8146(01)00174-1
  68. 68. Holstege DM, Puschner B, Whitehead G, Galey FD. Screening and mass spectral confirmation of β-lactam antibiotic residues in milk using LC-MS/MS. Journal of agricultural and food chemistry. 2002;50:406-411. DOI: 10.1021/jf010994s
  69. 69. Debayle D, Dessalces G, Grenier-Loustalot MF. Multi-residue analysis of traces of pesticides and antibiotics in honey by HPLC-MS-MS. Analytical and Bioanalytical Chemistry. 2008;391:1011-1020. DOI: 10.1007/s00216-008-2003-2
  70. 70. Denev P, Ciz M, Ambrozova G, Lojek A, Yanakieva I, Kratchanova M. Solid-phase extraction of berries’ anthocyanins and evaluation of their antioxidative properties. Food Chemistry. 2010;123:1055-1061. DOI: 10.1016/j.foodchem.2010.05.061
  71. 71. Chico J, Rúbies A, Centrich F, Companyó R, Prat MD, Granados M. High-throughput multiclass method for antibiotic residue analysis by liquid chromatography–tandem mass spectrometry. Journal of Chromatography A. 2008;1213:189-199. DOI: 10.1016/j.chroma.2008.10.023
  72. 72. Yamaguchi T, Okihashi M, Harada K, Uchida K, Konishi Y, Kajimura K, Hirata K, Yamamoto Y. Rapid and easy multiresidue method for the analysis of antibiotics in meats by ultrahigh-performance liquid chromatography–tandem mass spectrometry. Journal of agricultural and food chemistry. 2015;63:5133-5140. DOI: 10.1021/acs.jafc.5b00170
  73. 73. Tian H, Wang J, Zhang Y, Li S, Jiang J, Tao D, Zheng N. Quantitative multiresidue analysis of antibiotics in milk and milk powder by ultra-performance liquid chromatography coupled to tandem quadrupole mass spectrometry. Journal of chromatography B. 2016;1033:172-179. DOI: 10.1016/j.jchromb.2016.08.023
  74. 74. Renew JE, Huang CH. Simultaneous determination of fluoroquinolone, sulfonamide, and trimethoprim antibiotics in wastewater using tandem solid phase extraction and liquid chromatography–electrospray mass spectrometry. Journal of Chromatography A. 2004;1042:113-121. DOI: 10.1016/j.chroma.2004.05.056
  75. 75. Adrian J, Pinacho DG, Granier B, Diserens JM, Sánchez-Baeza F, Marco MP. A multianalyte ELISA for immunochemical screening of sulfonamide, fluoroquinolone and ß-lactam antibiotics in milk samples using class-selective bioreceptors. Analytical and bioanalytical chemistry. 2008;391:1703-1712. DOI: 10.1007/s00216-008-2106-9
  76. 76. Heller DN, Nochetto CB, Rummel NG, Thomas MH. Development of multiclass methods for drug residues in eggs: hydrophilic solid-phase extraction cleanup and liquid chromatography/tandem mass spectrometry analysis of tetracycline, fluoroquinolone, sulfonamide, and β-lactam residues. Journal of agricultural and food chemistry. 2006;54:5267-5278. DOI: 10.1021/jf0605502
  77. 77. Haller MY, Müller SR, McArdell CS, Alder AC, Suter MJ. Quantification of veterinary antibiotics (sulfonamides and trimethoprim) in animal manure by liquid chromatography–mass spectrometry. Journal of Chromatography A. 2002;952:111-120. DOI: 10.1016/S0021-9673(02)00083-3
  78. 78. Hirsch R, Ternes TA, Haberer K, Mehlich A, Ballwanz F, Kratz KL. Determination of antibiotics in different water compartments via liquid chromatography–electrospray tandem mass spectrometry. Journal of chromatography A. 1998;815:213-223. DOI: 10.1016/S0021-9673(98)00335-5
  79. 79. Hartig C, Storm T, Jekel M. Detection and identification of sulphonamide drugs in municipal waste water by liquid chromatography coupled with electrospray ionisation tandem mass spectrometry. Journal of Chromatography A. 1999;854:163-173. DOI: 10.1016/S0021-9673(99)00378-7
  80. 80. Bernal J, Ares AM, Pól J, Wiedmer SK. Hydrophilic interaction liquid chromatography in food analysis. Journal of Chromatography A. 2011;1218:7438-7452. DOI: 10.1016/j.chroma.2011.05.004
  81. 81. Tóth G, Panić-Janković T, Mitulović G. Pillar array columns for peptide separations in nanoscale reversed-phase chromatography. Journal of Chromatography A. 2019;1603:426-432. DOI: 10.1016/j.chroma.2019.06.067
  82. 82. Bens GA, Crombez E, Moerloose PD. Applications of a modified “Isohydric solvent system” in HPLC on silica gel for the analysis of the macrolide antibiotics turimycins and spiramycins. Journal of Liquid Chromatography. 1982 Jan 1;5(8):1449-1465. DOI: 10.1080/01483918208062843
  83. 83. Serrano JM, Silva M. Rapid and sensitive determination of aminoglycoside antibiotics in water samples using a strong cation-exchange chromatography non-derivatisation method with chemiluminescence detection. Journal of Chromatography A. 2006;1117:176-183. DOI: 10.1016/j.chroma.2006.03.086
  84. 84. Kim BH, Lee SC, Lee HJ, Ok JH. Reversed-phase liquid chromatographic method for the analysis of aminoglycoside antibiotics using pre-column derivatization with phenylisocyanate. Biomedical Chromatography. 2003;17:396-403. DOI: 10.1002/bmc.254
  85. 85. Kahsay G, Song H, Van Schepdael A, Cabooter D, Adams E. Hydrophilic interaction chromatography (HILIC) in the analysis of antibiotics. Journal of pharmaceutical and biomedical analysis. 2014;87:142-154. DOI: 10.1016/j.jpba.2013.04.015
  86. 86. Kostrzewa M, Sparbier K, Maier T, Schubert S. MALDI-TOF MS: an upcoming tool for rapid detection of antibiotic resistance in microorganisms. PROTEOMICS–Clinical Applications. 2013;7:767-778. DOI: 10.1002/prca.201300042
  87. 87. Adhikari AN, Gallagher RC, Wang Y, Currier RJ, Amatuni G, Bassaganyas L, Chen F, Kundu K, Kvale M, Mooney SD, Nussbaum RL: The role of exome sequencing in newborn screening for inborn errors of metabolism. Nature Medicine. 2020;10:1-6. DOI: 10.1038/s41591-020-0966-5
  88. 88. Vanessa Shaw. Dixon M, MacDonald A, White FJ. Disorders of Amino Acid Metabolism, Organic Acidaemias and Urea Cycle Disorders. In: Vanessa Shaw, editor. Clinical Paediatric Dietetics, 5th ed. Wiley-Blackwell; 2020. p. 513-598 DOI: 10.1002/9781119467205.ch28
  89. 89. Guthrie R, Susi A. A simple phenylalanine method for detecting phenylketonuria in large populations of newborn infants. Pediatrics. 1963;32:338-343
  90. 90. Céspedes N, Valencia A, Echeverry CA, Arce-Plata MI, Colón C, Castiñeiras DE, Hurtado PM, Cocho JA, Herrera S, Arévalo-Herrera M. Reference values of amino acids, acylcarnitines and succinylacetone by tandem mass spectrometry for use in newborn screening in southwest Colombia. Colombia Médica. 2017; 48:113-9. DOI: 10.25100/cm.v48i3.2180
  91. 91. Millington DS, Kodo N, Norwood DL, Roe CR. Tandem mass spectrometry: a new method for acylcarnitine profiling with potential for neonatal screening for inborn errors of metabolism. Journal of inherited metabolic disease. 1990;13:321-324. DOI: 10.1007/BF01799385
  92. 92. Chace DH, DiPerna JC, Naylor EW. Laboratory integration and utilization of tandem mass spectrometry in neonatal screening: a model for clinical mass spectrometry in the next millennium. Acta Paediatrica. 1999;88:45-47. DOI: 10.1111/j.1651-2227.1999.tb01156.x
  93. 93. Chen WH, Hsieh SL, Hsu KP, Chen HP, Su XY, Tseng YJ, Chien YH, Hwu WL, Lai F. Web-based newborn screening system for metabolic diseases: machine learning versus clinicians. Journal of medical Internet research. 2013;15:e98. DOI: 10.2196/jmir.2495
  94. 94. Hall PL, Marquardt G, McHugh DM, Currier RJ, Tang H, Stoway SD, Rinaldo P. Postanalytical tools improve performance of newborn screening by tandem mass spectrometry. Genetics in Medicine. 2014;16:889-895. DOI: 10.1038/gim.2014.62
  95. 95. la Marca G. Mass spectrometry in clinical chemistry: the case of newborn screening. Journal of pharmaceutical and biomedical analysis. 2014;101:174-182. DOI: 10.1016/j.jpba.2014.03.047
  96. 96. Clinical and Laboratory Standard Institute. Newborn screening by tandem mass spectrometry; Approved guideline. Wayne (PA): Clinical and Laboratory Standard Institute, 2010
  97. 97. Miller IV JH, Poston PA, Karnes HT. A quantitative method for acylcarnitines and amino acids using high resolution chromatography and tandem mass spectrometry in newborn screening dried blood spot analysis. Journal of Chromatography B. 2012;903:142-149. DOI: 10.1016/j.jchromb.2012.07.008
  98. 98. Wang C, Zhu H, Cai Z, Song F, Liu Z, Liu S. Newborn screening of phenylketonuria using direct analysis in real time (DART) mass spectrometry. Analytical and bioanalytical chemistry. 2013;405:3159-3164. DOI: 10.1007/s00216-013-6713-8
  99. 99. Chaimbault PE, Petritis KO, Elfakir CL, Dreux MI. Determination of 20 underivatized proteinic amino acids by ion-pairing chromatography and pneumatically assisted electrospray mass spectrometry. Journal of Chromatography A. 1999;855:191-202. DOI: 10.1016/S0021-9673(99)00685-8
  100. 100. Petritis K, Chaimbault P, Elfakir C, Dreux M. Parameter optimization for the analysis of underivatized protein amino acids by liquid chromatography and ionspray tandem mass spectrometry. Journal of Chromatography A. 2000;896:253-263. DOI: 10.1016/S0021-9673(00)00582-3
  101. 101. Piraud M, Vianey-Saban C, Petritis K, Elfakir C, Steghens JP, Morla A, Bouchu D. ESI-MS/MS analysis of underivatised amino acids: a new tool for the diagnosis of inherited disorders of amino acid metabolism. Fragmentation study of 79 molecules of biological interest in positive and negative ionisation mode. Rapid Communications in Mass Spectrometry. 2003;17:1297-1311. DOI: 10.1002/rcm.1054
  102. 102. Piraud M, Vianey-Saban C, Petritis K, Elfakir C, Steghens JP, Bouchu D. Ion-pairing reversed-phase liquid chromatography/electrospray ionization mass spectrometric analysis of 76 underivatized amino acids of biological interest: a new tool for the diagnosis of inherited disorders of amino acid metabolism. Rapid Communications in Mass Spectrometry: An International Journal Devoted to the Rapid Dissemination of Up-to-the-Minute Research in Mass Spectrometry. 2005;19:1587-602. DOI: 10.1002/rcm.1957
  103. 103. Zakaria R, Allen KJ, Koplin JJ, Roche P, Greaves RF. Advantages and challenges of dried blood spot analysis by mass spectrometry across the total testing process. Ejifcc. 2016;27:288
  104. 104. Chace DH, Kalas TA, Naylor EW. Use of tandem mass spectrometry for multianalyte screening of dried blood specimens from newborns. Clinical chemistry. 2003;49:1797-1817. DOI: 10.1373/clinchem.2003.022178
  105. 105. Fabie NA, Pappas KB, Feldman GL. The current state of newborn screening in the United States. Pediatric Clinics. 2019;66:369-386. DOI: 10.1016/j.pcl.2018.12.007
  106. 106. Wiley V, Carpenter K, Wilcken B. Newborn screening with tandem mass spectrometry: 12 months’ experience in NSW Australia. Acta Paediatrica. 1999;88:48-51.DOI: 10.1111/j.1651-2227.1999.tb01157.x
  107. 107. Pieragostino D, Cicalini I, Di Michele S, Fusilli P, Cotugno G, Ferrante R, Bucci I, Dionisi-Vici C, Stuppia L, De Laurenzi V, Rossi C. A Case of Suspected Hyperphenylalaninemia at Newborn Screening by Tandem Mass Spectrometry during Total Parenteral Nutrition. Metabolites. 2020;10:44. DOI: 10.3390/metabo10020044
  108. 108. Gouda AS, Nazim WS. Development of a simple method for the analysis of phenylalanine in dried blood spot using tandem mass spectrometry. Egyptian Journal of Medical Human Genetics. 2020;21(1):1-6
  109. 109. la Marca G, Malvagia S, Pasquini E, Innocenti M, Fernandez MR, Donati MA, Zammarchi E. The inclusion of succinylacetone as marker for tyrosinemia type I in expanded newborn screening programs. Rapid Communications in Mass Spectrometry: An International Journal Devoted to the Rapid Dissemination of Up-to-the-Minute Research in Mass Spectrometry. 2008;22(6):812-818
  110. 110. Oglesbee D, Sanders KA, Lacey JM, Magera MJ, Casetta B, Strauss KA, Tortorelli S, Rinaldo P, Matern D. Second-tier test for quantification of alloisoleucine and branched-chain amino acids in dried blood spots to improve newborn screening for maple syrup urine disease (MSUD). Clinical chemistry. 2008;54(3):542-549
  111. 111. Kim JI, Noh JY, Kim M, Park JM, Song HW, Kang MJ, Pyun JC. Newborn screening by matrix-assisted laser desorption/ionization mass spectrometry based on parylene-matrix chip. Analytical biochemistry. 2017 Aug 1;530:31-39
  112. 112. Yunus ZM, Rahman SA, Choy YS, Keng WT, Ngu LH. Pilot study of newborn screening of inborn error of metabolism using tandem mass spectrometry in Malaysia: outcome and challenges. Journal of Pediatric Endocrinology and Metabolism. 2016;29:1031-1039. DOI: 10.1515/jpem-2016-0028
  113. 113. Lim JS, Tan ES, John CM, Poh S, Yeo SJ, Ang JS, Adakalaisamy P, Rozalli RA, Hart C, Tan ET, Ranieri E. Inborn Error of Metabolism (IEM) screening in Singapore by electrospray ionization-tandem mass spectrometry (ESI/MS/MS): An 8 year journey from pilot to current program. Molecular genetics and metabolism. 2014;113(1-2):53-61
  114. 114. Filiano JJ, Bellimer SG, Kunz PL. Tandem mass spectrometry and newborn screening: pilot data and review. Pediatric Neurology 2002;26:201-204 DOI: 10.1016/s0887-8994(01)00384-8
  115. 115. Burlina AB, Polo G, Salviati L, Duro G, Zizzo C, Dardis A, Bembi B, Cazzorla C, Rubert L, Zordan R, Desnick RJ. Newborn screening for lysosomal storage disorders by tandem mass spectrometry in North East Italy. Journal of Inherited Metabolic Disease: Official Journal of the Society for the Study of Inborn Errors of Metabolism. 2018;41:209-219. DOI:10.1007/s10545-017-0098-3
  116. 116. GargU, Dasouki M. Expanded newborn screening of inherited metabolic disorders by tandem mass spectrometry: clinical and laboratory aspects. Clinical biochemistry. 2006;39:315-332. DOI: 10.1016/j.clinbiochem.2005.12.009
  117. 117. Mak CM, Lee HC, Chan AY, Lam CW. Inborn errors of metabolism and expanded newborn screening: review and update. Critical reviews in clinical laboratory sciences. 2013;50:142-162. DOI: 10.3109/10408363.2013.847896
  118. 118. Chace DH. Mass spectrometry in the clinical laboratory. Chemical reviews. 2001;101:445-478. DOI: 10.1021/cr990077+
  119. 119. Millington DS, Kodo N, Terada N, Roe D, Chace DH. The analysis of diagnostic markers of genetic disorders in human blood and urine using tandem mass spectrometry with liquid secondary ion mass spectrometry. International Journal of Mass Spectrometry and Ion Processes. 1991;111:211-228. DOI: 10.1016/0168-1176(91)85056-R
  120. 120. Shokri M, Karimi P, Zamanifar H, Kazemi F, Badfar G, Azami M. Phenylketonuria screening in Iranian newborns: a systematic review and meta-analysis. BMC pediatrics. 2020;20:1-6. DOI: 10.1186/s12887-020-02230-6
  121. 121. Shibata N, Hasegawa Y, Yamada K, Kobayashi H, Purevsuren J, Yang Y, Dung VC, Khanh NN, Verma IC, Bijarnia-Mahay S, Lee DH. Diversity in the incidence and spectrum of organic acidemias, fatty acid oxidation disorders, and amino acid disorders in Asian countries: selective screening vs. expanded newborn screening. Molecular Genetics and Metabolism Reports. 2018;16:5-10. DOI: 10.1016/j.ymgmr.2018.05.003
  122. 122. Piras D, Locci E, Palmas F, Ferino G, Fanos V, Noto A, D’aloja E, Finco G. Rare disease: a focus on metabolomics. Expert Opinion on Orphan Drugs. 2016;4:1229-1237. DOI: 10.1080/21678707.2016.1252671
  123. 123. Ozben T. Expanded newborn screening and confirmatory follow-up testing for inborn errors of metabolism detected by tandem mass spectrometry. Clinical Chemistry and Laboratory Medicine. 2013;51:157-176. DOI: 10.1515/cclm-2012-0472
  124. 124. Allard P, Grenier A, Korson MS, Zytkovicz TH. Newborn screening for hepatorenal tyrosinemia by tandem mass spectrometry: analysis of succinylacetone extracted from dried blood spots. Clinical biochemistry. 2004;37:1010-1015. DOI: 10.1016/j.clinbiochem.2004.07.006
  125. 125. Stinton C, Geppert J, Freeman K, Clarke A, Johnson S, Fraser H, Sutcliffe P, Taylor-Phillips S. Newborn screening for Tyrosinemia type 1 using succinylacetone–a systematic review of test accuracy. Orphanet journal of rare diseases. 2017;12:1-8. DOI: 10.1186/s13023-017-0599-z
  126. 126. Huemer, M., Kožich, V., Rinaldo, P., Baumgartner, M. R., Merinero, B., Pasquini, E., Ribes, A., & Blom, H. J. Newborn screening for homocystinurias and methylation disorders: systematic review and proposed guidelines. Journal of inherited metabolic disease, 2015;38:1007-1019. DOI: 10.1007/s10545-015-9830-z
  127. 127. Matern D, Tortorelli S, Oglesbee D, Gavrilov D, Rinaldo P. Reduction of the false-positive rate in newborn screening by implementation of MS/MS-based second-tier tests: the Mayo Clinic experience (2004-2007). Journal of Inherited Metabolic Disease. 2007;30:585-592. DOI:10.1007/s10545-007-0691-y
  128. 128. Hayden, Melvin R, and Suresh C Tyagi. “Homocysteine and reactive oxygen species in metabolic syndrome, type 2 diabetes mellitus, and atheroscleropathy: the pleiotropic effects of folate supplementation.” Nutrition journal 2004 vol. 3 4. 10 DOI:10.1186/1475-2891-3-4
  129. 129. Bonetti F, Brombo G, Zuliani G. The relationship between hyperhomocysteinemia and neurodegeneration. Neurodegenerative disease management. 2016;6:133-145. DOI: 10.2217/nmt-2015-0008
  130. 130. Hua Y, Zhao H, Kong Y, Ye M. Association between the MTHFR gene and Alzheimer’s disease: a meta-analysis. International Journal of Neuroscience. 2011;121:462-471. DOI:10.3109/00207454.2011.578778
  131. 131. Yi-Le Wu, Xiu-Xiu Ding, Ye-Huan Sun, Hui-Yun Yang, Liang Sun. Methylenetetrahydrofolate reductase (MTHFR) C677T/A1298C polymorphisms and susceptibility to Parkinson's disease: A meta-analysis. Journal of the Neurological Sciences. 2013; 335:14-21 DOI: 10.1016/j.jns.2013.09.006
  132. 132. Miletić J, Drakulić D, Pejić S, et al. Prooxidant-antioxidant balance, advanced oxidation protein products and lipid peroxidation in Serbian patients with Parkinson’s disease. International Journal of Neuroscience. 2018;128:600-607. DOI:10.1080/00207454.2017.1403916
  133. 133. J. Miletic, S. Pejic, A. Todorovic, A. Valenta Sоbot, D. Drakulic, I. Pavlovic, T. Ilic, A. Stefanovic, M. Prostran, M. Stojanov. Antioxidant status and clinicopathological parameters in patients with Parkinson's disease. Vojno sanitetski pregled. 2018 DOI: 10.2298/VSP180718148M
  134. 134. Miletic Vukajlovic J, Drakulić D, Pejić S, Ilić TV, Stefanović A, Petković M, Schiller J. Increased plasma phosphatidylcholine/lysophosphatidylcholine ratios in patients with Parkinson's disease. Rapid Communication in Mass Spectrometry. 2019 DOI: 10.1002/rcm.8595
  135. 135. Chace DH, Millington DS, Terada N, Kahler SG, Roe CR, Hofman LF. Rapid diagnosis of phenylketonuria by quantitative analysis for phenylalanine and tyrosine in neonatal blood spots by tandem mass spectrometry. Clinical Chemistry 1993;39:66-71
  136. 136. MacGloin H, Guilder L, Cleary M, Davison J, Uudelepp M, Chakrapani A. G262 Maple syrup urine disease (MSUD) referred to tertiary services before and after newborn screening (NBS). Archives of Disease in Childhood. 2019;104(Suppl 2):A106. DOI: 10.1136/archdischild-2019-rcpch.254
  137. 137. Rashed MS, Ozand PT, Bucknall MP, Little D. Diagnosis of inborn errors of metabolism from blood spots by acylcarnitines and amino acids profiling using automated electrospray tandem mass spectrometry. Pediatric research. 1995;38:324-331. DOI: 10.1203/00006450-199509000-00009
  138. 138. Stroek K, Boelen A, Bouva MJ, De Sain-van der Velden M, Schielen PC, Maase R, Engel H, Jakobs B, Kluijtmans LA, Mulder MF, Rubio-Gozalbo ME. Evaluation of 11 years of newborn screening for maple syrup urine disease in the Netherlands and a systematic review of the literature: Strategies for optimization. JIMD Reports. 2020 DOI: 10.1002/jmd2.12124
  139. 139. Chace DH, Hillman SL, Millington DS, Kahler SG, Roe CR, Naylor EW. Rapid diagnosis of maple syrup urine disease in blood spots from newborns by tandem mass spectrometry. Clinical Chemistry. 1995;41:62-68. DOI: 10.1093/clinchem/41.1.62
  140. 140. Heldt K, Schwahn B, Marquardt I, Grotzke M, Wendel U. Diagnosis of MSUD by newborn screening allows early intervention without extraneous detoxification. Molecular genetics and metabolism. 2005;84:313-316. DOI: 10.1016/j.ymgme.2004.11.010
  141. 141. Couce ML, Ramos F, Bueno MA, Díaz J, Meavilla S, Bóveda MD, Fernandez-Marmiesse A, Garcia-Cazorla A. Evolution of maple syrup urine disease in patients diagnosed by newborn screening versus late diagnosis. European Journal of Paediatric Neurology. 2015;19:652-659. DOI: 10.1016/j.ejpn.2015.07.009
  142. 142. Simon E, Fingerhut R, Baumkötter J, Konstantopoulou V, Ratschmann R, Wendel U. Maple syrup urine disease: favourable effect of early diagnosis by newborn screening on the neonatal course of the disease. Journal of Inherited Metabolic Disease: Official Journal of the Society for the Study of Inborn Errors of Metabolism. 2006;29:532-537. DOI: 10.1007/s10545-006-0315-y
  143. 143. Fingerhut R, Simon E, Maier EM, Hennermann JB, Wendel U. Maple syrup urine disease: newborn screening fails to discriminate between classic and variant forms. Clinical chemistry. 2008;54:1739-1741. DOI: 10.1373/clinchem.2008.105270
  144. 144. De Castro-Hamoy LG, Chiong MA, Estrada SC, Cordero CP. Challenges in the management of patients with maple syrup urine disease diagnosed by newborn screening in a developing country. Journal of community genetics. 2017;8:9-15. DOI: 10.1007/s12687-016-0281-5
  145. 145. Huang HP, Chu KL, Chien YH, Wei ML, Wu ST, Wang SF, Hwu WL. Tandem mass neonatal screening in Taiwan—report from one center. Journal of the Formosan Medical Association. 2006;105:882-886. DOI: 10.1016/S0929-6646(09)60173-X
  146. 146. Zytkovicz TH, Fitzgerald EF, Marsden D, Larson CA, Shih VE, Johnson DM, Strauss AW, Comeau AM, Eaton RB, Grady GF. Tandem mass spectrometric analysis for amino, organic, and fatty acid disorders in newborn dried blood spots: a two-year summary from the New England Newborn Screening Program. Clinical chemistry. 2001;47:1945-1955.DOI: 10.1093/clinchem/47.11.1945
  147. 147. Myers KA, Reeves M, Wei XC, Khan A. Cerebral edema in maple syrup urine disease despite newborn screening diagnosis and early initiation of treatment. JIMD Reports. 2012;3:103-106. DOI: 10.1007/8904_2011_69
  148. 148. Agadi S, Sutton VR, Quach MM, Riviello Jr JJ. The electroencephalogram in neonatal maple syrup urine disease: a case report. Clinical EEG and neuroscience. 2012;43:64-67. DOI: 10.1177/1550059411429521
  149. 149. Hassan FA, El-Mougy F, Sharaf SA, Mandour I, Morgan MF, Selim LA, Hassan SA, Salem F, Oraby A, Girgis MY, Mahmoud IG. Inborn errors of metabolism detectable by tandem mass spectrometry in Egypt: the first newborn screening pilot study. Journal of medical screening. 2016;23:124-129. DOI: 10.1177/0969141315618229
  150. 150. Jeong JS, Sim HJ, Lee YM, Yoon HR, Kwon HJ, Hong SP. Chromatographic diagnosis of maple syrup urine disease by measuring the L-alloisoleucine/L-phenylalanine ratio in dried blood spots. Journal of Chromatography B. 2011;879:2171-2174. DOI: 10.1016/j.jchromb.2011.05.023
  151. 151. Puckett RL, Lorey F, Rinaldo P, Lipson MH, Matern D, Sowa ME, Levine S, Chang R, Wang RY, Abdenur JE. Maple syrup urine disease: further evidence that newborn screening may fail to identify variant forms. Molecular genetics and metabolism. 2010;100:136-142. DOI: 10.1016/j.ymgme.2009.11.010
  152. 152. Eberlin LS, Dill AL, Costa AB, Ifa DR, Cheng L, Masterson T, et al. Cholesterol Sulfate Imaging in Human Prostate Cancer Tissue by Desorption Electrospray Ionization Mass Spectrometry. Analytical Chemistry. 2010;82(9):3430-3434
  153. 153. Eberlin LS, Norton I, Orringer D, Dunn IF, Liu X, Ide JL, et al. Ambient mass spectrometry for the intraoperative molecular diagnosis of human brain tumors. Proceedings of the National Academy of Sciences. 2013;110(5):1611-1616
  154. 154. Sans M, Zhang J, Lin JQ, Feider CL, Giese N, Breen MT, et al. Performance of the MasSpec Pen for Rapid Diagnosis of Ovarian Cancer. Clinical Chemistry. 2019;65(5):674-683
  155. 155. Torre LA, Trabert B, DeSantis CE, Miller KD, Samimi G, Runowicz CD, et al. Ovarian cancer statistics, 2018. CA Cancer J Clin. 2018;68(4):284-296
  156. 156. Siegel RL, Miller KD, Jemal A. Cancer statistics, 2018. CA Cancer J Clin. 2018;68(1):7-30
  157. 157. Brown HM, Pirro V, Cooks RG. From DESI to the MasSpec Pen: Ambient Ionization Mass Spectrometry for Tissue Analysis and Intrasurgical Cancer Diagnosis. Clinical Chemistry. 2018;64(4):628-630
  158. 158. Tzafetas M, Mitra A, Paraskevaidi M, Bodai Z, Kalliala I, Bowden S, et al. The intelligent knife (iKnife) and its intraoperative diagnostic advantage for the treatment of cervical disease. Proceedings of the National Academy of Sciences. 2020;117(13):7338-7346
  159. 159. St John ER, Balog J, McKenzie JS, Rossi M, Covington A, Muirhead L, et al. Rapid evaporative ionisation mass spectrometry of electrosurgical vapours for the identification of breast pathology: towards an intelligent knife for breast cancer surgery. Breast Cancer Res. 2017;19(1):59
  160. 160. Santagata S, Eberlin LS, Norton I, Calligaris D, Feldman DR, Ide JL, Liu X, Wiley JS, Vestal ML, Ramkissoon SH, Orringer DA. Intraoperative mass spectrometry mapping of an onco-metabolite to guide brain tumor surgery. Proceedings of the National Academy of Sciences. 2014;111(30):11121-11126
  161. 161. Calligaris D, Norton I, Feldman DR, Ide JL, Dunn IF, Eberlin LS, Graham Cooks R, Jolesz FA, Golby AJ, Santagata S, Agar NY. Mass spectrometry imaging as a tool for surgical decision-making. Journal of Mass Spectrometry. 2013;48(11):1178-87
  162. 162. Basu SS, Regan MS, Randall EC, Abdelmoula WM, Clark AR, Lopez BG, Cornett DS, Haase A, Santagata S, Agar NY. Rapid MALDI mass spectrometry imaging for surgical pathology. NPJ precision oncology. 2019;3(1):1-5

Written By

Jadranka Miletić Vukajlović and Tanja Panić-Janković

Submitted: 03 June 2020 Reviewed: 19 February 2021 Published: 08 April 2021