Next Article in Journal
LASP1, CERS6, and Actin Form a Ternary Complex That Promotes Cancer Cell Migration
Previous Article in Journal
Re-Shaping the Pancreatic Cancer Tumor Microenvironment: A New Role for the Metastasis Suppressor NDRG1
Previous Article in Special Issue
Small and Large Extracellular Vesicles Derived from Pleural Mesothelioma Cell Lines Offer Biomarker Potential
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Biomarker Reproducibility Challenge: A Review of Non-Nucleotide Biomarker Discovery Protocols from Body Fluids in Breast Cancer Diagnosis

by
Fatemeh Safari
1,
Cheka Kehelpannala
2,3,†,
Azadeh Safarchi
1,4,†,
Amani M. Batarseh
2,3,* and
Fatemeh Vafaee
1,5,6,*
1
School of Biotechnology and Biomolecular Sciences, University of New South Wales (UNSW Sydney), Sydney, NSW 2052, Australia
2
BCAL Diagnostics Ltd., Suite 506, 50 Clarence St, Sydney, NSW 2000, Australia
3
BCAL Dx, The University of Sydney, Sydney Knowledge Hub, Merewether Building, Sydney, NSW 2006, Australia
4
Microbiomes for One Systems Health, Health and Biosecurity, CSIRO, Westmead, NSW 2145, Australia
5
UNSW Data Science Hub (uDASH), University of New South Wales (UNSW Sydney), Sydney, NSW 2052, Australia
6
OmniOmics.ai Pty Ltd., Sydney, NSW 2035, Australia
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
Cancers 2023, 15(10), 2780; https://doi.org/10.3390/cancers15102780
Submission received: 24 February 2023 / Revised: 2 May 2023 / Accepted: 10 May 2023 / Published: 16 May 2023
(This article belongs to the Special Issue Decoding Cancer Signals Using Liquid Biopsy)

Abstract

:

Simple Summary

Various studies and techniques have been designed to discover biofluid-derived biomarkers for non-invasive early detection and prognosis of cancers. Despite the importance of non-invasive biomarker discovery in cancer diagnosis and management, the reported markers are often inconsistent and irreproducible across different studies and cohorts. In this article, we reviewed the ongoing trend of non-nucleotide biomarkers, including lipidomics, proteomics and metabolomics, derived from body fluids, with a focus on breast cancer, and reviewed the inconstancies in the biomarker discovery pipelines across pre-analytical, analytical, and post-analytical phases, covering the diversity of approaches from sample processing to predictive modelling and validation.

Abstract

Breast cancer has now become the most commonly diagnosed cancer, accounting for one in eight cancer diagnoses worldwide. Non-invasive diagnostic biomarkers and associated tests are superlative candidates to complement or improve current approaches for screening, early diagnosis, or prognosis of breast cancer. Biomarkers detected from body fluids such as blood (serum/plasma), urine, saliva, nipple aspiration fluid, and tears can detect breast cancer at its early stages in a minimally invasive way. The advancements in high-throughput molecular profiling (omics) technologies have opened an unprecedented opportunity for unbiased biomarker detection. However, the irreproducibility of biomarkers and discrepancies of reported markers have remained a major roadblock to clinical implementation, demanding the investigation of contributing factors and the development of standardised biomarker discovery pipelines. A typical biomarker discovery workflow includes pre-analytical, analytical, and post-analytical phases, from sample collection to model development. Variations introduced during these steps impact the data quality and the reproducibility of the findings. Here, we present a comprehensive review of methodological variations in biomarker discovery studies in breast cancer, with a focus on non-nucleotide biomarkers (i.e., proteins, lipids, and metabolites), highlighting the pre-analytical to post-analytical variables, which may affect the accurate identification of biomarkers from body fluids.

1. Background

The number of women diagnosed with breast cancer is increasing every year [1]. Female breast cancer surpassed lung cancer as the most diagnosed cancer in the world in 2020, with approximately 2.3 million new cases diagnosed [2]. Although statistics show that the mortality rate of breast cancer patients is low compared to lung, colorectal, liver, and stomach cancer patients [2], the breast cancer burden is costly and an enormous obstacle to increasing the quality of life for women and girls around the world [1].
One of the key approaches to breast cancer management and control is cost-effective screening and early detection [3]. Although mammography is a widely used breast cancer screening technology, it has technical, logistic, and diagnostic limitations with respect to false positives and negatives, convenience and participation, limitations with younger women and dense breasts, exposure to radiation, and the limitation of detecting tumours of small sizes in the early stage of breast cancer [4,5]. Therefore, alternative tools for the screening and detection of breast cancer are urgently needed.
Over the past decade, there has been a surge of interest in the study of metabolite, protein, and lipid biomarkers derived from high-throughput molecular profiling of various biofluids of patients with breast cancer, such as blood (serum/plasma) [6,7,8], urine [9,10,11], saliva [12,13], ductal lavage fluid [14], nipple aspiration fluid [14,15,16], and tears [17,18]. While increasing evidence from numerous studies corroborates the utility of non-nucleotide biomarkers in diagnosing breast cancer, there have often been inconsistencies in biomarkers reported by different studies. One of the major contributors to the observed inconsistencies is the lack of gold-standard methods and protocols across different phases of biomarker discovery and laboratory testing, including the pre-analytical phase (specimen collection and sample processing), analytical phase (measurement of analytes), and post-analytical phase (data pre-processing, statistical analysis, model development, interpretation of results, and reporting) [19,20].
There are several factors across each of these phases which can impact the outcomes and the reproducibility of the findings, that is, the ability to replicate the same results for the same condition [21]. It is, therefore, essential to review diverse approaches commonly practised in each phase of liquid biopsy biomarker discovery to help research and industry sectors to adopt the best practices and standardise their pipelines. There has been a dearth of comprehensive reviews on experimental and methodological variations from the pre-analytical to post-analytical phases of liquid biopsies for breast cancer diagnosis and beyond. Furthermore, while former studies have reviewed liquid biopsies in breast cancer [22,23,24,25], the focus has often been on circulating tumour cells (CTCs) or nucleotide-based biomarkers (e.g., circulating tumour DNA (ctDNA), circulating or extracellular vesicle-encapsulated microRNA, and platelet-derived RNAs), with non-nucleotide biomarkers often limited to proteins. Therefore, non-nucleotide-based circulating biomarkers have been reviewed to a much less extent, demanding focused attention given the differences in the pre-analytical and analytical phases and diversity of techniques of molecular profiling for non-nucleotide compared to nucleotide-based analytes.
To address this resource gap, we conducted a comprehensive search of the literature published in the last two decades (2001–2023), focusing on high-throughput omics approaches in breast cancer liquid biopsy using Google Scholar, PubMed, Elsevier, and Scopus databases to query a combination of medical subject headings (MeSH) and terms including breast cancer, proteomics, metabolomics, lipidomics, liquid biopsy, plasma, serum, blood, urine, saliva, tear, nipple aspiration fluid, and ductal lavage fluid. We extracted over 200 relevant public shed articles (Supplementary Table S1), reviewed their biomarker discovery pipelines, and investigated the trend in the target biofluids and biomarker types. Figure 1 illustrates different factors affecting biomarker discovery outcomes from the pre-analytical to post-analytical phase, according to the findings of previous studies. More details on exemplar studies are summarised in Table 1 (and Supplementary Table S2) to showcase the diversity of protocols adopted across different phases of biomarker discovery, focusing on circulating non-nucleotide-based breast cancer diagnostic makers (lipids, proteins, and metabolites), highlighting the demand for standardised procedures.

2. Pre-Analytical Variables

In biomarker discovery studies, body fluid sources, sample collection procedures, handling, preparation steps, and storage conditions are defined as pre-analytical variables [42,43]. They are one of the most error-prone, time-consuming, and laborious steps in biomarker identification, and they affect the sensitivity, reproducibility, and selectivity of analysis and need to be carefully considered during the project design [44]. In the following subsections, we will outline the intricacy of the pre-analytical phase and its significance for biomarker discovery.

2.1. Biofluids Are Excellent Sources of Biomarkers

Recently, different types of body fluids have obtained great attention as sources of biomarkers for the detection and monitoring of breast cancer due to their low complexity and simpler sample collection and processing procedures compared to solid tissues, sustainable accessibility, and the ability to be measured repeatably in a minimally invasive way [45]. The major challenge in biomarker discovery from body fluids is the identification of biomarkers specific to the type of cancer. For example, a proteomic analysis of five different body fluids by Zhao et al. suggested that the proteome of body fluids may indicate the holistic functions of the whole body rather than that of adjacent tissues [46]. Therefore, the identification of biomarkers released into the body fluid by cancerous lesions may be difficult. Nonetheless, the metabolic changes that occur in the body due to the onset of cancer can be reflected in the metabolic/proteomic profile of body fluids. Furthermore, daily water intake or microbiome profile may alter the protein or metabolite concentration in a patient’s body fluid [47,48] and the biomarker concentration may depend on the sample collection method. Thus, the pre-analytical phase of biomarker discovery workflows should be stringently standardised.
The selection of appropriate body fluids depends on the type of omics study (i.e., proteomics, lipidomics, or metabolomics), as one specimen may be advantageous over the other. For example, urine samples, mainly composed of metabolites and end products of biochemical reactions, are more suitable for metabolomic analysis [49]. Furthermore, compared to saliva, which comprises 99% water and 0.3% protein, serum and plasma are more appropriate for proteomic investigations [50]. In the following sections, we discuss the commonly used biofluids for biomarker discovery.

2.1.1. Serum and Plasma

Blood is believed to have the most complex human-derived circulating biomarkers and therefore has attracted considerable research attention. So far, over 12,000 proteins, 600 lipids, and 300 metabolites have been profiled from blood samples [51,52,53], and the concentrations of many circulating analytes were found to be different in plasma and serum [54,55,56]. For example, Liu et al. revealed that some metabolites, including most amino acids, hypoxanthine, carbohydrates, b-hydroxybutyrate, and glycerol-3-phosphate, were significantly lower in plasma compared to serum. In contrast, other metabolic products such as citrate, fumarate, pyruvate, glycerate, nitrogen metabolites, urate, and hydroxylamine were significantly higher in the plasma [54]. Furthermore, studies indicate that the total concentration of several lipids, including triglycerides (TGs), phosphatidylcholines (PCs), and HDL cholesterol, were higher in serum than in EDTA or citrate plasma [57,58].
Breier et al. reported that the reliability of metabolite measurements was slightly higher in serum samples compared to plasma [59]. The reason for this may be the higher metabolite concentration in serum compared to plasma, which provides greater sensitivity for biomarker identification [60,61]. However, the concentration of some metabolites involved in platelet aggregation will be different from their actual level in serum as the clotting process causes these metabolite levels to increase. Therefore, such metabolites will need to be measured from plasma [58]. In the study by Ishikawa et al., it has been demonstrated that plasma is more suitable than serum for studying lipid biomarkers because the clotting process was found to affect serum lipid levels [62]. Moreover, lipids showed the lowest biological variation in plasma citrate samples, implying the suitability of plasma for quantitative targeted lipidomics [60]. Nonetheless, the method and conditions by which the plasma was prepared need to be standardised to avoid detecting differences due to the time used, temperature or type of tubes, centrifuge used, or how the sample is stored (e.g., 4 °C −20 °C, −80 °C, or snap frozen).
When the blood clot is removed during serum preparation, the concentration of high-abundance circulating proteins, such as fibrinogen, will significantly decrease in serum, making it much easier to detect low-abundance proteins. At the same time, some proteins are released from the platelet during the blood coagulation process. This phenomenon can vary sample-to-sample and may lead to the false positive identification of protein biomarkers from serum [63,64]. A study by Tammen et al. suggested citrate plasma or platelet-depleted EDTA plasma for studying the low-molecular-weight proteome [65]. In 2005, the HUPO’s Human Plasma Proteome Project (HPPP) recommended using EDTA plasma as the preferred sample for all proteomic analyses [66]. Therefore, it is not possible to measure the biomarkers of interest from plasma and serum interchangeably. Based on the aims of the study and the target biomarker, either plasma or serum may need to be chosen.
As shown in Figure 2, a greater tendency to use serum over plasma has not been observed in breast cancer metabolomics investigations. The number of metabolomics studies that used serum as the biofluid sample of choice was relatively similar to those that utilised plasma samples. However, plasma was the preferred matrix over serum for breast cancer lipidomic investigations, with approximately 60% of the publications reporting on plasma as opposed to approximately 24% reporting on serum. In contrast, serum samples were used in approximately 44% of the studies focusing on proteomics investigations of breast cancer, which is much higher than plasma selection.

2.1.2. Urine

Urine is one of the most widely used human body fluids for routine testing due to its less complex composition [67,68]. Many studies on urine biomarkers for breast cancer screening and diagnosis are still in the discovery phase; hence, further cohort investigations are needed to validate their sensitivity and specificity [22].
There are several types of urine collection approaches, including random, first-morning, second-morning, and 24-h collections [69]. Each kind has unique advantages and disadvantages for metabolomic, proteomic, and lipidomic investigations. Although a random urine sample is presumably the most straightforward collection approach, it is rarely the preferred choice, as depending on the collection time, urine may be excessively diluted due to water intake, and the patient’s diet and exercise would have affected its composition [68,70]. The first-morning urine sample is generally considered appropriate for proteomic studies because it contains the largest amount of total proteins [71] and shows the lowest variation compared to the 24-h urine samples [68,72]. Conversely, the midstream second-morning urine collected after an overnight fast is recommended for metabolomic profiling, as the pattern of metabolites in the first-morning urine may reflect nutrients consumed the day before [69,73]. Although urine collection time is a critical factor, it has been neglected by many studies focused on urinary metabolomics in breast cancer [74,75,76]. However, in a few investigations, it has been indicated that first-morning urine collection was used [77,78].
In terms of lipidomic analysis, there is a lack of information demonstrating the characteristics of each urine sample type based on the time of sampling for lipid biomarker discovery. Furthermore, few investigations have been performed on urinary lipidomics in patients with breast cancer, in which the detailed information of urine collection protocols has not been addressed well [79,80].
Another aspect to consider using urine as the source of biomarkers is the difference in the microbiome composition of the urinary tract and the vaginal tract in women. Due to the microbiome–host interaction, the results can be affected. The microbiota may produce and secrete proteins, lipids, etc., which may confound the biomarker discovery and may also metabolise the host-secreted biomarkers in the sample. It has been shown that urinary microbiota composition differs by menopausal status in patients with breast cancer [81]. Moreover, regardless of menopausal status, cancerous patients had increased levels of Gram-positive bacteria, including Corynebacterium, Staphylococcus, Actinomyces, and Propionibacteriaceae [81], which may influence the metabolite and protein content of urine.

2.1.3. Tears

The tear’s composition, especially proteins, can be substantially affected due to the sample collection procedure [82,83,84]. Schirmer’s test strips (STSs) and microcapillary tubes (MCTs) are the most popular tear sampling procedures [85]. Pieragostino et al. [86] reviewed the advantages and disadvantages of collection techniques previously. STSs have been used in most proteomics studies in breast cancer so far [17,18,87]. Results from the analysis by Nättinen et al. [83] indicated that Schirmer strip samples had a ten-fold greater mean total protein content compared to MCTs. To date, there is no agreement on how the tear sampling procedures impact the proteomic data. Sample handling, such as strip cutting, has been shown to increase the risk of contamination and protein loss, making the results even more variable [84]. Therefore, the most appropriate and reliable tear sampling approaches are needed for the accurate and repeatable detection of tear biomarkers.

2.1.4. Nipple Aspiration Fluid

Nipple aspirate fluid (NAF) in non-lactating women is a fluid secreted by breast epithelial duct cells and can be collected with various degrees of effectiveness, ranging from 34% to 90% by utilising a milk-expressing pump, nasal oxytocin spray, and gentle breast massage [31,88,89,90]. Proteins are the main components of NAF, with concentrations up to 170 mg/mL, which can be more than that found in plasma [91]. However, there are some challenges when using NAF as a source of protein biomarkers. Firstly, NAF droplets may not be acquired from the duct where carcinogenesis has occurred [92]. Furthermore, it has been shown that the colour and viscosity of NAF can affect biomarker identification when using spectrophotometry approaches [92]. Li et al. proposed that the notable differences in the results spectra between NAF samples in a group may stem from several reasons, including the biological variation in the breast duct’s microenvironment and variability of the protein concentration in the samples (equal sample volume was examined rather than equal protein concentration) [14]. Given these variations and challenges in NAF sample examinations, it is difficult to cross-compare the findings of different investigations. Another challenge in using NAF is that the microbiome profile and host–microbiome interaction may interfere with the biomarker studies. The study by Chan et al. showed that the microbiota composition of NAF significantly differs in patients with breast cancer compared to healthy women, which may affect the multi-omics profile of NAF [93] for biomarker discovery.

2.1.5. Saliva

The safe, non-invasive, and repeatable collection makes saliva a good target for biomarker discovery. Investigations showed significant differences in the level of metabolites in saliva that can be used as biomarkers for breast cancer diagnosis [37,94,95]. However, the exceptionally diverse composition of saliva arising from age, diet, gender, and time of day of the collection makes it a challenging choice of biofluid for biomarker studies.
Protein degradation is one of the main reasons for the irreproducibility of salivary proteomic analyses. The proteolytic degradation commences just as the proteins enter the oral cavity and continues post-collection of salivary samples, leading to substantial differences in biomarker profiles [70]. Furthermore, salivary biomarkers can be affected by the site of collection. For example, Cui et al. showed that the concentration of several metabolites was different in whole saliva, parotid saliva, and submandibular/sublingual saliva [96]. Moreover, Assad et al. propounded that small variations in the collection and storage procedure affect the free amino acid content of saliva as it comprises proteinases and peptidases [97], resulting in irreproducible results between studies.

2.1.6. Extracellular Vesicles

Extracellular vesicles (EVs) are rich sources of circulating biomarkers in blood that have been of interest in many recent studies, with demonstrated utility in breast cancer diagnosis, as reviewed previously [98]. Continuous production, release, and uptake of existing EVs by different types of blood cells, as well as the delay between blood collection and preparation of plasma or serum, need to be considered when EVs are used for biomarker discovery [99,100]. It is shown that physical activity undertaken prior to sample collection, besides other pre-analytical parameters such as collection tube, centrifugation, and storage time, may influence morphology, size, and stability, as well as the downstream characterisation of EVs [101,102]. EV isolation and enrichment are other discriminatory pre-analytical factors in many studies, as there is no established gold-standard protocol to purify and isolate EVs. For instance, centrifugation is one of the main parameters that impact the reproducibility of EV isolation and purification [102,103]. This may complicate cross-comparison between studies as well as the external validation of biomarkers. This lack of standardised guidelines in EV research has triggered international efforts and consortiums, such as EV-TRACK (https://evtrack.org/index.php, accessed on 2 May 2023), to facilitate the standardisation of EV research through increased systematic reporting [104].
Based on the study published in 2020 [105], the concentration and size of the microvesicles (MVs), which are a sub-type of EVs, differ in plasma and serum. While MVs have lower concentrations in serum, small-sized MVs are higher in serum than large-sized MVs. In another study by Palviainen et al., the protein profiles of plasma EVs were different between serum and plasma [106]. In order to reduce vesicle release from blood cells, most procedures suggest using plasma rather than serum [101]. EVs and MVs in cancer biomarker discovery have previously been reviewed in detail [99,101,107,108]. In breast cancer studies focusing on EVs, plasma was used as the main source compared to serum [108], regardless of the type of EV composition.

2.2. Sample Collection and Processing Variables Impact the Discovery of Accurate Biomarkers

In addition to biofluid type, other pre-analytical variables, including anti-coagulants, collection tubes, incubation times (pre-centrifugation processing delay), storage time and temperature, and freeze–thaw cycles, can also influence biomarker levels, thereby affecting the analytical reproducibility [42,43,109]. Some distinguished influential variables that may occur during sample collection and handling are presented in Table 2 to highlight the importance of considering these facets in prospective proteomic, metabolomic, and lipidomic studies. The information presented in Table 2 reiterates that the pre-analytical phase should be meticulously controlled and regulated to prevent unfavourable impacts on biomarker discovery and underscores the need for highly standardised protocols.

2.3. Trends in Non-Invasive, Non-Nucleotide Biomarker Discovery for Breast Cancer

As discussed above, in biomarker discovery studies, the ease of sample collection, reproducibility, and effective variables are some of the critical factors. Biomarker investigations for the detection and prognosis of breast cancer are more concentrated on non-invasive approaches rather than a tissue biopsy. Figure 2 illustrates the proportion of metabolomic, lipidomic, and proteomic investigations carried out on various biofluid samples of breast cancer between January 2001 and April 2023. It demonstrates that the number of studies exploiting non-nucleotide-based biomarkers from various biofluids has increased in the last ten years. Although proteomics has dominated the field for many years, there has been a shift to metabolomics and lipidomics since 2015. Regarding biofluid sources, although various biofluids have been exploited for biomarker discovery, blood continues to be the primary biofluid for biomarker discovery (plasma and serum). Notably, serum was the primary source before 2015, and plasma was the primary source from 2015 to 2019. The preference for choosing blood over other biofluids might be due to the fact that, compared to the other biofluids, fewer variables, including exposure to air, possible effects of their microbiome on the abundance and composition of analytes, time of collection, and the high proportion analytes related to adjacent tissues may affect the study outcomes [137,138,139,140].
Furthermore, protocols and analysis pipelines of plasma and serum may be more standardised compared to other biofluids. Proteomics and metabolomics are emerging fields that have expanded rapidly as a result of parallel improvements in bioanalytical platforms and methods for data analysis [141]. As shown in Figure 2, the trend of research using proteomics to identify biomarkers has been overtaken by lipidomics and metabolomics in more recent years. This may be due to the development of new protocols and methods for metabolome and lipidome purification, advances in analytical techniques, and awareness of their potential use for biomarker discovery.

3. Analytical Techniques for Biomarker Discovery

Apart from the pre-analytical variables, the wide dynamic ranges, sensitivity, and specificity of analytical methods are major challenges in biomarkers discovery, which can affect the reproducibility of biomarker identification. For example, because some biomarkers have a very low abundance in the selected biofluid, the sensitivity of the analytical method can limit the number of discovered proteins [70]. Detailed information on commonly used techniques in proteomic, metabolomic, and lipidomic investigations is included in Table 3 and summarised below.

3.1. Proteomic Approaches

Proteomic workflows can be categorised as gel-based and gel-free methods coupled with array-based and mass spectrometry-based (MS) techniques [159]. Mass-spectrometry (MS) is the most commonly used approach in proteomic studies of breast cancer [160]. Time-of-flight, triple quadrupole, and orbitrap mass spectrometers can be coupled with different ionisation procedures, including surface-enhanced laser desorption/ionisation (SELDI), matrix-assisted laser desorption/ionisation (MALDI), and electrospray ionisation (ESI) for proteomic applications [160]. Although most of the investigations utilised the SELDI-TOF-MS method for breast cancer diagnosis as a potential discovery method, the reproducibility was questionable due to the low resolution of SELDI-TOF-MS data and chip-to-chip variation. In contrast, MALDI-TOF-MS shows higher reliability and robustness and is favoured in clinical proteomics [161]. However, it is not without limitations; for example, MALDI-TOF-MS is sensitive to impurities such as salt, causing problems with the reproducibility of the results [68].
Two-dimensional gel electrophoresis (2-DE) is a technique widely used in qualitative proteomic investigations of breast cancer [10,28]. However, this technique has some drawbacks, including weak inter-assay reproducibility, low sensitivity for the detection of proteins with either very low PH (<3) or high PH (>10) values, and too small (<10 kD) or too large (>150 kD) molecular masses, as well as the inability to identify hydrophobic and low abundant proteins [162]. In contrast, the two-dimensional difference in the gel electrophoresis (2D-DIGE) approach has demonstrated higher sensitivity and improved reproducibility [155].
Other factors, such as diversity in binding/washing buffer conditions and the chemistry of ProteinChip surfaces, can influence the binding and identification of various proteins, leading to discrepancies in biomarker discovery [27]. For example, IMAC3 (Immobilized Metal Affinity Capture) chips capture proteins via chelation of metal ions, whereas H4 chips absorb by hydrophobic interaction; consequently, the proteins captured by these chips are distinct and would lead to irreproducible results [163,164]. Therefore, analytical procedures should be standardised among research and clinical laboratories for a precise interpretation and interlaboratory comparison of data.

3.2. Metabolomic Approaches

Two main analytical techniques are commonly employed in metabolomic investigations: mass spectrometry and nuclear magnetic resonance (NMR) spectroscopy [32]. Although NMR has the capability to measure metabolites with high reproducibility in complex samples without the need for pre-preparation of biological fluids, it shows low sensitivity [165]. Mass spectrometry techniques used for breast cancer studies include ultra-high performance liquid chromatography coupled with quadrupole time-of-flight (UPLC-QTOF-MS) [166,167], gas chromatography-mass spectrometry (GC-MS) [168,169,170], liquid chromatography-mass spectrometry (LC-MS) [8,171], and ultra-fast liquid chromatography-tandem mass spectrometry (UFLC-MS/MS) [172]. However, the LC-MS and GC-MS methods have been frequently applied for biofluids [173]. LC-MS stands as the most suitable approach for the sensitive identification of biomolecules with high reproducibility [174], while GC-MS shows relatively stronger chromatography with distinct peak separation [175].

3.3. Lipidomic Approaches

Technological advancements in liquid chromatography, high-resolution accurate mass spectrometry, and NMR spectroscopy have improved the high throughput analysis of lipid molecules [176]. Many mass-spectrometry-based approaches are used in lipidomic studies, each with unique characteristics, advantages, and disadvantages [177]. Mass spectrometry imaging (MSI), direct infusion or shotgun MS, and MS accompanied by initial chromatographic separation such as GC, LC, and thin-layer chromatography (TLC) are the main three infrastructures of lipidomic investigations [147]. Shotgun MS, in which the analyte is not separated by prior chromatography, performs poorly in detecting less-ionisable and low-abundant lipids due to ion suppression, during which the signals stemming from weakly ionised lipid species are buried in the signal of strongly ionised lipids [178,179]. However, the detection of such lipids can be improved by a pre-separation approach, such as LC-MS, which has demonstrated high sensitivity, specificity, and remarkable separation efficiency for lipids [147].

4. Post-Analytical Steps and Variations

4.1. Data Pre-Processing

Mass spectrometry-based techniques have become the mainstream methods for high-throughput and unbiased proteomics, metabolomics, and lipidomics profiling. Several forms of proprietary and open-source software have been developed for data acquisition and quantification, as discussed elsewhere [180,181]. These tools have different underlying assumptions and algorithms for searching (e.g., database vs. de novo) and molecular species quantification [182], which contributes to the discrepancy of generated data across different studies. A comprehensive benchmarking is required to compare data acquisition and quantification techniques and to provide a guideline for the best practices.
Once quantified, high-throughput spectrometry or spectroscopy data are often subject to multiple pre-processing steps to stabilise variance, reduce systematic bias or technical variations, and impute missing data. The choice of pre-processing approach can substantially affect the data quality and validity of downstream analyses. For instance, Mertens [183] argued in favour of log-transformation to mitigate the skewness and standardise spectrometry data, which has raised concerns regarding using so-called “closure normalisation”, e.g., data normalised by the sum of the combined expression in exerting spurious biases in the correlations between the spectral measures masking true population associations. Nonetheless, the diversity of the available pre-processing statistical approaches demands benchmarking studies to systematically investigate their effect on the quality of data and the reproducibility of the biomarkers identified. Välikangas et al. [184], for instance, evaluated normalisation methods in quantitative label-free proteomics and demonstrated the variations in outcomes of downstream analyses (e.g., differential expression) depending on the choice of the normalisation method. Despite the importance of pre-processing, we frequently observed unclear and incomplete descriptions of the approaches undertaken in the literature we have reviewed in relation to the non-nucleotide biomarkers of breast cancer (Table 1 and Supplementary Table S2).

4.2. Biomarker Signature Panel Identification (Feature Selection)

From the computational perspective, signature panel identification can be formulated as a feature selection or extraction problem, which implies the selection of a set of molecules (e.g., proteins, lipids, or metabolites) that best stratify the groups of interest (e.g., cancer vs. control) or the extraction of latent features from the entire omics profile (e.g., embeddings derived via dimensionality reduction). Feature selection has been historically performed via differential analysis (i.e., statistical hypothesis tests such as t-test or Mann–Whitney U test). However, while differential analysis can detect functionally relevant molecules, it is ineffective in selecting features with optimal predictive power [185] as it is a univariate approach overlooking nonlinear relationships among multiple biomarkers, whose collective effect contributes to the prediction of a phenotype, disease outcome, or treatment response. Several sophisticated machine learning-based methods have been developed by the computer science community for feature extraction or selection of predictive variables from high-dimensional data, which can substantially enhance signature panel identification, and the development of predictive models and cancer diagnostics as previously benchmarked [186]. Despite the proven utility of machine learning and nonlinear, multivariate feature selection in identifying biomarker signatures with high sensitivity and specificity, statistical hypotheses testing has been the dominant approach adopted in non-nucleotide breast cancer biomarker discovery, as outlined in Supplementary Table S2.

4.3. Biomarker Predictive Modelling (Classification)

After feature selection (or extraction), the identified biomarker signature panel can be used as predictive variables of a classifier algorithm to stratify patients into categories of interest (e.g., cancer vs. normal). A classifier algorithm often implements a mathematical function that maps input data to a category upon learning from a training cohort. Different classifiers have been implemented as multi-variate cancer diagnostics models, including commonly used algorithms such as random forest, support vector machines, logistic regression, artificial neural networks, and ensemble approaches (i.e., predictive models composed of a weighted combination of multiple classifiers) [187]. For a long time, improving the prediction accuracy has been the primary focus of biomarker discovery predictive modelling. However, biomarker discovery methods should be assessed based on prediction accuracy as well as robustness, defined as the generalisability of the model to diverse cohorts. In recent years, the stability of biomarker discovery has gained more attention, as reviewed previously [188]. Nonetheless, in breast cancer liquid biopsy studies, the adoption of classifiers as diagnostic models has been limited (Supplementary Table S2), contributing to the lack of highly predictive and robust diagnostic tests.

4.4. Clinical Validation

Extensive validation is necessary before the clinical implementation of a diagnostic test. Validation of a predictive model using the dataset at hand (referred to as the development dataset) is often referred to as an internal validation, wherein the dataset is divided into the test and train sets, using the latter for model development and optimisation and the former for model validation. In addition, to mitigate model overfitting, particularly in small datasets, data re-sampling techniques, such as bootstrapping or cross-validation, can be used to account for the selection bias and to quantify the stability of the predictive performance [189].
Based on our literature review, the majority of breast cancer liquid biopsy studies have only reported the prediction performance of biomarkers upon internal validation, which is not sufficient to confirm model generalisability. In order to progress towards implementation and technology readiness, extensive external validation is required, wherein the model’s predictive performance is quantified using data collected from participant cohorts external, temporally and/or geographically, to the development dataset [189].
Besides the validation of the prediction models, the analytical parameters should be optimised, followed by the validation of the parameters according to regulatory guidelines [190,191]. The clinical performance of the test should then be compared to the gold-standard method, e.g., mammography [192,193]. When the technology is implemented, prospective clinical studies should be conducted to assess if the assay improves patient outcomes and reduces healthcare costs [192,194].

5. Conclusions and Future Perspective

Our major biofluid biomarker discovery pathway throughout the last decades was focused mainly on nucleotide-based biomarkers for early breast cancer diagnosis. However, in recent years, the investigation of proteomics, lipidomics, metabolomics, and microbiome profiles, along with EV cargo, has been increased to introduce new biomarker profiles, not only for blood but also for other types of body fluids, as we have comprehensively reviewed here. We also reviewed the effect of different procedures, from sample collection and processing to data analysis and validation. The lack of standard protocols in different parts of biomarker discovery can be a key factor hindering the clinical implementation and manufacturing of commercialisable assays or clinical tests. Therefore, one of the future efforts in breast cancer biomarker studies is to standardise the liquid biopsy assay procedures and analysis platforms. This will give a better opportunity to combine and compare results from different studies and develop breast cancer liquid biopsy consortiums to advance and validate liquid biopsy technologies, homogenise guidelines, and standardise data for the development of breast cancer biomarkers. Some initiatives have already been implemented by the National Institute of Health (https://prevention.cancer.gov/major-programs/liquid-biopsy-consortium, accessed on 2 May 2023), targeted for early-stage cancer detection on a wide range of cancer types.
Due to the ongoing advances in non-invasive biomarker discovery, technology, and data analytics, the future of the field is moving towards multi-omics liquid biopsy and non-invasive blood tests (or other bodily fluids) through the simultaneous assessment of different omics data (e.g., genomics, transcriptomics, and proteomics) from body fluids for cancer detection and monitoring. Multi-omics approaches could provide complementary information on the presence of the dysregulated bodily processes leading to disease, enabling early detection of tumours, and they have demonstrated utility in enhancing the sensitivity and specificity of cancer detection as we construct a fuller picture [195]. Despite its advantages, multi-omics liquid biopsy is facing slow adoption and implementation. So far, there have been limited studies using this approach for breast cancer identification emerging over the last few years (Supplementary Table S1). One major obstacle is limited sample availability and/or technical difficulties associated with generating complete multi-omics datasets due to the uneven maturity of different omics approaches. Moreover, the growing gap between generating large volumes of data compared to data processing capacity and available integrated datasets are of concern. Additional efforts are needed for the standardisation of multi-omics operational procedures and data integration, from robust pre-processing and operational guidelines to data integration and validation.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/cancers15102780/s1, Supplementary Table S1: The titles of metabolomic, lipidomic, proteomic, and multi-omics investigations carried out on various biofluid samples of breast cancer between January 2001 and April 2023; Supplementary Table S2: The details of the approaches undertaken in relation to the discovery of non-nucleotide biomarkers in breast cancer investigations (selected studies).

Author Contributions

Conceptualization, F.V., A.M.B. and A.S.; methodology, F.S., F.V., C.K. and A.S.; formal analysis, F.S., F.V. and A.S.; investigation, F.S., F.V. and A.S.; data curation, F.S., F.V., A.S. and C.K.; writing—original draft preparation, F.S., F.V., A.S. and C.K.; writing—review and editing, F.S., F.V., A.S. and A.M.B.; visualization, F.S., F.V. and A.S.; supervision, F.V. and A.M.B.; funding acquisition, F.V. All authors have read and agreed to the published version of the manuscript.

Funding

UNSW Industry Network Seed Funding (INSF) 2021–2022.

Data Availability Statement

All data generated or analysed during this study are included either as a supplementary file or are publicly available and properly referenced in the manuscript.

Acknowledgments

We acknowledge the Industry Network Seed Funding (INSF) provided by the University of New South Wales (UNSW Sydney), Faculty of Science.

Conflicts of Interest

A.B. declares a relationship with BCAL Diagnostics, and F.V. declares a relationship with OmniOmics.ai Pty Ltd. Otherwise, the authors declare no conflict of interest.

References

  1. Perry, S.; Kowalski, T.L.; Chang, C.H. Quality of Life Assessment in Women with Breast Cancer: Benefits, Acceptability and Utilization. Health Qual. Life Outcomes 2007, 5, 24. [Google Scholar] [CrossRef] [PubMed]
  2. Sung, H.; Ferlay, J.; Siegel, R.L.; Laversanne, M.; Soerjomataram, I.; Jemal, A.; Bray, F. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J. Clin. 2021, 71, 209–249. [Google Scholar] [CrossRef] [PubMed]
  3. Sharma, R. Global, Regional, National Burden of Breast Cancer in 185 Countries: Evidence from GLOBOCAN 2018. Breast Cancer Res. Treat. 2021, 187, 557–567. [Google Scholar] [CrossRef]
  4. Autier, P.; Boniol, M. Mammography Screening: A Major Issue in Medicine. Eur. J. Cancer 2018, 90, 34–62. [Google Scholar] [CrossRef]
  5. Løberg, M.; Lousdal, M.L.; Bretthauer, M.; Kalager, M. Benefits and Harms of Mammography Screening. Breast Cancer Res. 2015, 17, 63. [Google Scholar] [CrossRef] [PubMed]
  6. Bartkowiak, K.; Heidrich, I.; Kwiatkowski, M.; Banys-Paluchowski, M.; Andreas, A.; Wurlitzer, M.; Geffken, M.; Voß, H.; Zeller, T.; Blankenberg, S.; et al. Circulating Cellular Communication Network Factor 1 Protein as a Sensitive Liquid Biopsy Marker for Early Detection of Breast Cancer. Clin. Chem. 2022, 68, 344–353. [Google Scholar] [CrossRef] [PubMed]
  7. Orlandi, R.; De Bortoli, M.; Ciniselli, C.M.; Vaghi, E.; Caccia, D.; Garrisi, V.; Pizzamiglio, S.; Veneroni, S.; Bonini, C.; Agresti, R.; et al. Hepcidin and Ferritin Blood Level as Noninvasive Tools for Predicting Breast Cancer. Ann. Oncol. 2014, 25, 352–357. [Google Scholar] [CrossRef]
  8. Jasbi, P.; Wang, D.; Cheng, S.L.; Fei, Q.; Cui, J.Y.; Liu, L.; Wei, Y.; Raftery, D.; Gu, H. Breast Cancer Detection Using Targeted Plasma Metabolomics. J. Chromatogr. B Analyt. Technol. Biomed. Life Sci. 2019, 1105, 26–37. [Google Scholar] [CrossRef]
  9. Beretov, J.; Wasinger, V.C.; Millar, E.K.A.; Schwartz, P.; Graham, P.H.; Li, Y. Proteomic Analysis of Urine to Identify Breast Cancer Biomarker Candidates Using a Label-Free LC-MS/MS Approach. PLoS ONE 2015, 10, e0141876. [Google Scholar] [CrossRef]
  10. Gajbhiye, A.; Dabhi, R.; Taunk, K.; Vannuruswamy, G.; RoyChoudhury, S.; Adhav, R.; Seal, S.; Mane, A.; Bayatigeri, S.; Santra, M.K.; et al. Urinary Proteome Alterations in HER2 Enriched Breast Cancer Revealed by Multipronged Quantitative Proteomics. Proteomics 2016, 16, 2403–2418. [Google Scholar] [CrossRef]
  11. Soydinc, H.O.; Duranyildiz, D.; Guney, N.; Derin, D.; Yasasever, V. Utility of Serum and Urine Upar Levels for Diagnosis of Breast Cancer. Asian Pac. J. Cancer Prev. 2012, 13, 2887–2889. [Google Scholar] [CrossRef] [PubMed]
  12. Takayama, T.; Tsutsui, H.; Shimizu, I.; Toyama, T.; Yoshimoto, N.; Endo, Y.; Inoue, K.; Todoroki, K.; Min, J.Z.; Mizuno, H.; et al. Diagnostic Approach to Breast Cancer Patients Based on Target Metabolomics in Saliva by Liquid Chromatography with Tandem Mass Spectrometry. Clin. Chim. Acta 2016, 452, 18–26. [Google Scholar] [CrossRef] [PubMed]
  13. Castagnola, M.; Picciotti, P.M.; Messana, I.; Fanali, C.; Fiorita, A.; Cabras, T.; Calò, L.; Pisano, E.; Passali, G.C.; Iavarone, F.; et al. Potential Applications of Human Saliva as Diagnostic Fluid. Acta Otorhinolaryngol. Ital. 2011, 31, 347–357. [Google Scholar] [PubMed]
  14. Li, J.; Zhao, J.; Yu, X.; Lange, J.; Kuerer, H.; Krishnamurthy, S.; Schilling, E.; Khan, S.A.; Sukumar, S.; Chan, D.W. Identification of Biomarkers for Breast Cancer in Nipple Aspiration and Ductal Lavage Fluid. Clin. Cancer Res. 2005, 11, 8312–8320. [Google Scholar] [CrossRef] [PubMed]
  15. Noble, J.L.; Dua, R.S.; Coulton, G.R.; Isacke, C.M.; Gui, G.P.H. A Comparative Proteinomic Analysis of Nipple Aspiration Fluid from Healthy Women and Women with Breast Cancer. Eur. J. Cancer 2007, 43, 2315–2320. [Google Scholar] [CrossRef] [PubMed]
  16. Pawlik, T.M.; Fritsche, H.; Coombes, K.R.; Xiao, L.; Krishnamurthy, S.; Hunt, K.K.; Pusztai, L.; Chen, J.N.; Clarke, C.H.; Arun, B.; et al. Significant Differences in Nipple Aspirate Fluid Protein Expression between Healthy Women and Those with Breast Cancer Demonstrated by Time-of-Flight Mass Spectrometry. Breast Cancer Res. Treat. 2005, 89, 149–157. [Google Scholar] [CrossRef]
  17. Lebrecht, A.; Boehm, D.; Schmidt, M.; Koelbl, H.; Schwirz, R.L.; Grus, F.H. Diagnosis of Breast Cancer by Tear Proteomic Pattern. Cancer Genom. Proteom. 2009, 6, 177–182. [Google Scholar]
  18. Böhm, D.; Keller, K.; Pieter, J.; Boehm, N.; Wolters, D.; Siggelkow, W.; Lebrecht, A.; Schmidt, M.; Kölbl, H.; Pfeiffer, N.; et al. Comparison of Tear Protein Levels in Breast Cancer Patients and Healthy Controls Using a de Novo Proteomic Approach. Oncol. Rep. 2012, 28, 429–438. [Google Scholar] [CrossRef]
  19. Tiwari, E.; Pallipady, A.; Mishra, S. Pre-analytical, Analytical and Postanalytical Errors in Chemical Laboratory. Int. J. Sci. Res. 2013, 4, 2279–2281. [Google Scholar]
  20. Klont, F.; Horvatovich, P.; Govorukhina, N.; Bischoff, R. Pre- and Post-Analytical Factors in Biomarker Discovery. In Methods in Molecular Biology; Humana Press: New York, NY, USA, 2019; Volume 1959, pp. 1–22. [Google Scholar]
  21. Zhuang, W.; Camacho, L.; Silva, C.S.; Hong, H. Reproducibility Challenges for Biomarker Detection with Uncertain but Informative Experimental Data. Biomark Med. 2020, 14, 1255–1263. [Google Scholar] [CrossRef]
  22. Li, J.; Guan, X.; Fan, Z.; Ching, L.M.; Li, Y.; Wang, X.; Cao, W.M.; Liu, D.X. Non-Invasive Biomarkers for Early Detection of Breast Cancer. Cancers 2020, 12, 2767. [Google Scholar] [CrossRef] [PubMed]
  23. Li, D.; Lai, W.; Fan, D.; Fang, Q. Protein Biomarkers in Breast Cancer-Derived Extracellular Vesicles for Use in Liquid Biopsies. Am. J. Physiol. Cell Physiol. 2021, 321, C779–C797. [Google Scholar] [CrossRef] [PubMed]
  24. Wu, H.J.; Chu, P.Y. Current and Developing Liquid Biopsy Techniques for Breast Cancer. Cancers 2022, 14, 2052. [Google Scholar] [CrossRef]
  25. Seale, K.N.; Tkaczuk, K.H.R. Circulating Biomarkers in Breast Cancer. Clin. Breast Cancer 2022, 22, e319–e331. [Google Scholar] [CrossRef] [PubMed]
  26. Sun, Y.; Zhang, J.; Guo, F.; Zhao, W.; Zhan, Y.; Liu, C.; Fan, Y.; Wang, J. Identification of Apolipoprotein C-I Peptides as a Potential Biomarker and Its Biological Roles in Breast Cancer. Med. Sci. Monit. 2016, 22, 1152–1160. [Google Scholar] [CrossRef]
  27. Böhm, D.; Keller, K.; Wehrwein, N.; Lebrecht, A.; Schmidt, M.; Kölbl, H.; Grus, F.H. Serum Proteome Profiling of Primary Breast Cancer Indicates a Specific Biomarker Profile. Oncol. Rep. 2011, 26, 1051–1056. [Google Scholar] [CrossRef]
  28. Scumaci, D.; Tammè, L.; Fiumara, C.V.; Pappaianni, G.; Concolino, A.; Leone, E.; Faniello, M.C.; Quaresima, B.; Ricevuto, E.; Costanzo, F.S.; et al. Plasma Proteomic Profiling in Hereditary Breast Cancer Reveals a BRCA1-Specific Signature: Diagnostic and Functional Implications. PLoS ONE 2015, 10, e0129762. [Google Scholar] [CrossRef]
  29. Chen, I.H.; Xue, L.; Hsu, C.C.; Paez, J.S.P.; Panb, L.; Andaluz, H.; Wendt, M.K.; Iliuk, A.B.; Zhu, J.K.; Tao, W.A. Phosphoproteins in Extracellular Vesicles as Candidate Markers for Breast Cancer. Proc. Natl. Acad. Sci. USA 2017, 114, 3175–3180. [Google Scholar] [CrossRef]
  30. Corrêa, S.; Panis, C.; Binato, R.; Herrera, A.C.; Pizzatti, L.; Abdelhay, E. Identifying Potential Markers in Breast Cancer Subtypes Using Plasma Label-Free Proteomics. J. Proteom. 2017, 151, 33–42. [Google Scholar] [CrossRef]
  31. George, A.L.; Shaheed, S.U.; Sutton, C.W. High-throughput Proteomic Profiling of Nipple Aspirate Fluid from Breast Cancer Patients Compared with Non-cancer Controls: A Step Closer to Clinical Feasibility. J. Clin. Med. 2021, 10, 2243. [Google Scholar] [CrossRef]
  32. Park, J.; Shin, Y.; Kim, T.H.; Kim, D.H.; Lee, A. Plasma Metabolites as Possible Biomarkers for Diagnosis of Breast Cancer. PLoS ONE 2019, 14, e0225129. [Google Scholar] [CrossRef] [PubMed]
  33. Wei, Y.; Jasbi, P.; Shi, X.; Turner, C.; Hrovat, J.; Liu, L.; Rabena, Y.; Porter, P.; Gu, H. Early Breast Cancer Detection Using Untargeted and Targeted Metabolomics. J. Proteome Res. 2021, 20, 3124–3133. [Google Scholar] [CrossRef] [PubMed]
  34. Eniu, D.T.; Romanciuc, F.; Moraru, C.; Goidescu, I.; Eniu, D.; Staicu, A.; Rachieriu, C.; Buiga, R.; Socaciu, C. The Decrease of Some Serum Free Amino Acids Can Predict Breast Cancer Diagnosis and Progression. Scand. J. Clin. Lab. Investig. 2019, 79, 17–24. [Google Scholar] [CrossRef] [PubMed]
  35. Cala, M.; Aldana, J.; Sánchez, J.; Guio, J.; Meesters, R.J.W. Urinary Metabolite and Lipid Alterations in Colombian Hispanic Women with Breast Cancer: A Pilot Study. J. Pharm. Biomed. Anal. 2018, 152, 234–241. [Google Scholar] [CrossRef]
  36. Murata, T.; Yanagisawa, T.; Kurihara, T.; Kaneko, M.; Ota, S.; Enomoto, A.; Tomita, M.; Sugimoto, M.; Sunamura, M.; Hayashida, T.; et al. Salivary Metabolomics with Alternative Decision Tree-Based Machine Learning Methods for Breast Cancer Discrimination. Breast Cancer Res. Treat. 2019, 177, 591–601. [Google Scholar] [CrossRef]
  37. Zhong, L.; Cheng, F.; Lu, X.; Duan, Y.; Wang, X. Untargeted Saliva Metabonomics Study of Breast Cancer Based on Ultra Performance Liquid Chromatography Coupled to Mass Spectrometry with HILIC and RPLC Separations. Talanta 2016, 158, 351–360. [Google Scholar] [CrossRef]
  38. Jiang, N.; Zhang, G.; Pan, L.; Yan, C.; Zhang, L.; Weng, Y.; Wang, W.; Chen, X.; Yang, G. Potential Plasma Lipid Biomarkers in Early-Stage Breast Cancer. Biotechnol. Lett. 2017, 39, 1657–1666. [Google Scholar] [CrossRef]
  39. Chen, X.; Chen, H.; Dai, M.; Ai, J.; Li, Y.; Mahon, B.; Dai, S.; Deng, Y. Plasma Lipidomics Profiling Identified Lipid Biomarkers in Distinguishing Early-Stage Breast Cancer from Benign Lesions. Oncotarget 2016, 7, 36622–36631. [Google Scholar] [CrossRef]
  40. Gumà, J.; Adriá-Cebrián, J.; Ruiz-Aguado, B.; Albacar, C.; Girona, J.; Rodríguez-Calvo, R.; Martínez-Micaelo, N.; Lam, E.W.F.; Masana, L.; Guaita-Esteruelas, S. Altered Serum Metabolic Profile Assessed by Advanced 1h-Nmr in Breast Cancer Patients. Cancers 2021, 13, 4281. [Google Scholar] [CrossRef]
  41. Bel’Skaya, L.V.; Sarf, E.A.; Kosenok, V.K. Analysis of Saliva Lipids in Breast and Prostate Cancer by IR Spectroscopy. Diagnostics 2021, 11, 1325. [Google Scholar] [CrossRef]
  42. Tuli, L.; Ressom, H.W. LC-MS Based Detection of Differential Protein Expression. J. Proteom. Bioinform. 2009, 2, 416–438. [Google Scholar] [CrossRef] [PubMed]
  43. Ahmed, F.E. Sample Preparation and Fractionation for Proteome Analysis and Cancer Biomarker Discovery by Mass Spectrometry. J. Sep. Sci. 2009, 32, 771–798. [Google Scholar] [CrossRef] [PubMed]
  44. Luque-Garcia, J.L.; Neubert, T.A. Sample Preparation for Serum/Plasma Profiling and Biomarker Identification by Mass Spectrometry. J. Chromatogr. A 2007, 1153, 259–276. [Google Scholar] [CrossRef]
  45. Urabe, F.; Kosaka, N.; Ito, K.; Kimura, T.; Egawa, S.; Ochiya, T. Extracellular Vesicles as Biomarkers and Therapeutic Targets for Cancer. Am. J. Physiol. Cell Physiol. 2020, 318, C29–C39. [Google Scholar] [CrossRef]
  46. Zhao, M.; Yang, Y.; Guo, Z.; Shao, C.; Sun, H.; Zhang, Y.; Sun, Y.; Liu, Y.; Song, Y.; Zhang, L.; et al. A Comparative Proteomics Analysis of Five Body Fluids: Plasma, Urine, Cerebrospinal Fluid, Amniotic Fluid, and Saliva. Proteom. Clin. Appl. 2018, 12, e1800008. [Google Scholar] [CrossRef] [PubMed]
  47. Athanasatou, A.; Kandyliari, A.; Malisova, O.; Kapsokefalou, M. Fluctuation of Water Intake and of Hydration Indices during the Day in a Sample of Healthy Greek Adults. Nutrients 2019, 11, 793. [Google Scholar] [CrossRef] [PubMed]
  48. Katsani, K.R.; Sakellari, D. Saliva Proteomics Updates in Biomedicine. J. Biol. Res. 2019, 26, 17. [Google Scholar] [CrossRef]
  49. Günther, U.L. Metabolomics Biomarkers for Breast Cancer. Pathobiology 2015, 82, 153–165. [Google Scholar] [CrossRef]
  50. Kaczor-Urbanowicz, K.E.; Martin Carreras-Presas, C.; Aro, K.; Tu, M.; Garcia-Godoy, F.; Wong, D.T.W. Saliva Diagnostics—Current Views and Directions. Exp. Biol. Med. 2017, 242, 459–472. [Google Scholar] [CrossRef]
  51. Nanjappa, V.; Thomas, J.K.; Marimuthu, A.; Muthusamy, B.; Radhakrishnan, A.; Sharma, R.; Ahmad Khan, A.; Balakrishnan, L.; Sahasrabuddhe, N.A.; Kumar, S.; et al. Plasma Proteome Database as a Resource for Proteomics Research: 2014 Update. Nucleic Acids Res. 2013, 42, D959–D965. [Google Scholar] [CrossRef]
  52. Quehenberger, O.; Armando, A.M.; Brown, A.H.; Milne, S.B.; Myers, D.S.; Merrill, A.H.; Bandyopadhyay, S.; Jones, K.N.; Kelly, S.; Shaner, R.L.; et al. Lipidomics Reveals a Remarkable Diversity of Lipids in Human Plasma1. J. Lipid Res. 2010, 51, 3299–3305. [Google Scholar] [CrossRef] [PubMed]
  53. Lawton, K.A.; Berger, A.; Mitchell, M.; Milgram, K.E.; Evans, A.M.; Guo, L.; Hanson, R.W.; Kalhan, S.C.; Ryals, J.A.; Milburn, M.V. Analysis of the Adult Human Plasma Metabolome. Pharmacogenomics 2008, 9, 383–397. [Google Scholar] [CrossRef] [PubMed]
  54. Liu, L.; Aa, J.; Wang, G.; Yan, B.; Zhang, Y.; Wang, X.; Zhao, C.; Cao, B.; Shi, J.; Li, M.; et al. Differences in Metabolite Profile between Blood Plasma and Serum. Anal. Biochem. 2010, 406, 105–112. [Google Scholar] [CrossRef]
  55. Boyanton, B.L.; Blick, K.E. Stability Studies of Twenty-Four Analytes in Human Plasma and Serum. Clin. Chem. 2002, 48, 2242–2247. [Google Scholar] [CrossRef]
  56. Haymond, R.E.; Knight, J.A. Venous Serum, Capillary Serum, and Capillary Plasma Compared for Use in Determination of Lactate Dehydrogenase and Aspartate Aminotransferase Activities. Clin. Chem. 1975, 21, 896–897. [Google Scholar] [CrossRef] [PubMed]
  57. Hyötyläinen, T.; Orešič, M. Optimizing the Lipidomics Workflow for Clinical Studies—Practical Considerations. Anal. Bioanal. Chem. 2015, 407, 4973–4993. [Google Scholar] [CrossRef] [PubMed]
  58. Lima-Oliveira, G.; Monneret, D.; Guerber, F.; Guidi, G.C. Sample Management for Clinical Biochemistry Assays: Are Serum and Plasma Interchangeable Specimens? Crit. Rev. Clin. Lab. Sci. 2018, 55, 480–500. [Google Scholar] [CrossRef]
  59. Breier, M.; Wahl, S.; Prehn, C.; Fugmann, M.; Ferrari, U.; Weise, M.; Banning, F.; Seissler, J.; Grallert, H.; Adamski, J.; et al. Targeted Metabolomics Identifies Reliable and Stable Metabolites in Human Serum and Plasma Samples. PLoS ONE 2014, 9, e89728. [Google Scholar] [CrossRef]
  60. Paglia, G.; Del Greco, F.M.; Sigurdsson, B.B.; Rainer, J.; Volani, C.; Hicks, A.A.; Pramstaller, P.P.; Smarason, S.V. Influence of Collection Tubes during Quantitative Targeted Metabolomics Studies in Human Blood Samples. Clin. Chim. Acta 2018, 486, 320–328. [Google Scholar] [CrossRef]
  61. Yu, Z.; Kastenmüller, G.; He, Y.; Belcredi, P.; Möller, G.; Prehn, C.; Mendes, J.; Wahl, S.; Roemisch-Margl, W.; Ceglarek, U.; et al. Differences between Human Plasma and Serum Metabolite Profiles. PLoS ONE 2011, 6, e21230. [Google Scholar] [CrossRef]
  62. Ishikawa, M.; Maekawa, K.; Saito, K.; Senoo, Y.; Urata, M.; Murayama, M.; Tajima, Y.; Kumagai, Y.; Saito, Y. Plasma and Serum Lipidomics of Healthy White Adults Shows Characteristic Profiles by Subjects’ Gender and Age. PLoS ONE 2014, 9, e91806. [Google Scholar] [CrossRef] [PubMed]
  63. Ignjatovic, V.; Geyer, P.E.; Palaniappan, K.K.; Chaaban, J.E.; Omenn, G.S.; Baker, M.S.; Deutsch, E.W.; Schwenk, J.M. Mass Spectrometry-Based Plasma Proteomics: Considerations from Sample Collection to Achieving Translational Data. J. Proteome Res. 2019, 18, 4085–4097. [Google Scholar] [CrossRef] [PubMed]
  64. Geyer, P.E.; Voytik, E.; Treit, P.V.; Doll, S.; Kleinhempel, A.; Niu, L.; Müller, J.B.; Buchholtz, M.; Bader, J.M.; Teupser, D.; et al. Plasma Proteome Profiling to Detect and Avoid Sample-related Biases in Biomarker Studies. EMBO Mol. Med. 2019, 11, e10427. [Google Scholar] [CrossRef]
  65. Tammen, H.; Schulte, I.; Hess, R.; Menzel, C.; Kellmann, M.; Mohring, T.; Schulz-Knappe, P. Peptidomic Analysis of Human Blood Specimens: Comparison between Plasma Specimens and Serum by Differential Peptide Display. Proteomics 2005, 5, 3414–3422. [Google Scholar] [CrossRef] [PubMed]
  66. Omenn, G.S.; States, D.J.; Adamski, M.; Blackwell, T.W.; Menon, R.; Hermjakob, H.; Apweiler, R.; Haab, B.B.; Simpson, R.J.; Eddes, J.S.; et al. Overview of the HUPO Plasma Proteome Project: Results from the Pilot Phase with 35 Collaborating Laboratories and Multiple Analytical Groups, Generating a Core Dataset of 3020 Proteins and a Publicly-Available Database. Proteomics 2005, 5, 3226–3245. [Google Scholar] [CrossRef]
  67. Thongboonkerd, V. Urinary Proteomics: Towards Biomarker Discovery, Diagnostics and Prognostics. Mol. Biosyst. 2008, 4, 810–815. [Google Scholar] [CrossRef]
  68. Beretov, J.; Wasinger, V.C.; Graham, P.H.; Millar, E.K.; Kearsley, J.H.; Li, Y. Proteomics for Breast Cancer Urine Biomarkers. Adv. Clin. Chem. 2014, 63, 123–167. [Google Scholar] [PubMed]
  69. Lehmann, R. From Bedside to Bench—Practical Considerations to Avoid Pre-Analytical Pitfalls and Assess Sample Quality for High-Resolution Metabolomics and Lipidomics Analyses of Body Fluids. Anal. Bioanal. Chem. 2021, 413, 5567–5585. [Google Scholar] [CrossRef] [PubMed]
  70. Bauça, J.M.; Martínez-Morillo, E.; Diamandis, E.P. Peptidomics of Urine and Other Biofluids for Cancer Diagnostics. Clin. Chem. 2014, 60, 1052–1061. [Google Scholar] [CrossRef]
  71. Thongboonkerd, V.; Chutipongtanate, S.; Kanlaya, R. Systematic Evaluation of Sample Preparation Methods for Gel-Based Human Urinary Proteomics: Quantity, Quality, and Variability. J. Proteome Res. 2006, 5, 183–191. [Google Scholar] [CrossRef] [PubMed]
  72. Eric Thomas, C.; Sexton, W.; Benson, K.; Sutphen, R.; Koomen, J. Urine Collection and Processing for Protein Biomarker Discovery and Quantification. Cancer Epidemiol. Biomark. Prev. 2010, 19, 953–959. [Google Scholar] [CrossRef] [PubMed]
  73. Liu, X.; Yin, P.; Shao, Y.; Wang, Z.; Wang, B.; Lehmann, R.; Xu, G. Which Is the Urine Sample Material of Choice for Metabolomics-Driven Biomarker Studies? Anal. Chim. Acta 2020, 1105, 120–127. [Google Scholar] [CrossRef] [PubMed]
  74. Slupsky, C.M.; Steed, H.; Wells, T.H.; Dabbs, K.; Schepansky, A.; Capstick, V.; Faught, W.; Sawyer, M.B. Urine Metabolite Analysis Offers Potential Early Diagnosis of Ovarian and Breast Cancers. Clin. Cancer Res. 2010, 16, 5835–5841. [Google Scholar] [CrossRef] [PubMed]
  75. Lee, J.; Woo, H.M.; Kong, G.; Nam, S.J.; Chung, B.C. Discovery of Urinary Biomarkers in Patients with Breast Cancer Based on Metabolomics. Mass Spectrom. Lett. 2013, 4, 59–66. [Google Scholar] [CrossRef]
  76. More, T.H.; Taware, R.; Taunk, K.; Chanukuppa, V.; Naik, V.; Mane, A.; Rapole, S. Investigation of Altered Urinary Metabolomic Profiles of Invasive Ductal Carcinoma of Breast Using Targeted and Untargeted Approaches. Metabolomics 2018, 14, 107. [Google Scholar] [CrossRef]
  77. Silva, C.L.; Passos, M.; Câmara, J.S. Solid Phase Microextraction, Mass Spectrometry and Metabolomic Approaches for Detection of Potential Urinary Cancer Biomarkers—A Powerful Strategy for Breast Cancer Diagnosis. Talanta 2012, 89, 360–368. [Google Scholar] [CrossRef]
  78. Zhou, J.; Wang, Y.; Zhang, X. Metabonomics Studies on Serum and Urine of Patients with Breast Cancer Using 1H-NMR Spectroscopy. Oncotarget 2017, 5. [Google Scholar] [CrossRef]
  79. Kim, H.; Min, H.K.; Kong, G.; Moon, M.H. Quantitative Analysis of Phosphatidylcholines and Phosphatidylethanolamines in Urine of Patients with Breast Cancer by Nanoflow Liquid Chromatography/Tandem Mass Spectrometry. Anal. Bioanal. Chem. 2009, 393, 1649–1656. [Google Scholar] [CrossRef]
  80. Min, H.K.; Kong, G.; Moon, M.H. Quantitative Analysis of Urinary Phospholipids Found in Patients with Breast Cancer by Nanoflow Liquid Chromatography-Tandem Mass Spectrometry: II. Negative Ion Mode Analysis of Four Phospholipid Classes. Anal. Bioanal. Chem. 2010, 396, 1273–1280. [Google Scholar] [CrossRef]
  81. Wang, H.; Altemus, J.; Niazi, F.; Green, H.; Calhoun, B.C.; Sturgis, C.; Grobmyer, S.R.; Eng, C. Breast Tissue, Oral and Urinary Microbiomes in Breast Cancer. Oncotarget 2017, 8, 88122–88138. [Google Scholar] [CrossRef]
  82. Rentka, A.; Koroskenyi, K.; Harsfalvi, J.; Szekanecz, Z.; Szucs, G.; Szodoray, P.; Kemeny-Beke, A. Evaluation of Commonly Used Tear Sampling Methods and Their Relevance in Subsequent Biochemical Analysis. Ann. Clin. Biochem. 2017, 54, 521–529. [Google Scholar] [CrossRef]
  83. Nättinen, J.; Aapola, U.; Jylhä, A.; Vaajanen, A.; Uusitalo, H. Comparison of Capillary and Schirmer Strip Tear Fluid Sampling Methods Using Swath-Ms Proteomics Approach. Transl. Vis. Sci. Technol. 2020, 9, 16. [Google Scholar] [CrossRef] [PubMed]
  84. Ponzini, E.; Santambrogio, C.; De Palma, A.; Mauri, P.; Tavazzi, S.; Grandori, R. Mass Spectrometry-Based Tear Proteomics for Noninvasive Biomarker Discovery. Mass Spectrom. Rev. 2022, 41, 842–860. [Google Scholar] [CrossRef] [PubMed]
  85. Zhou, L.; Beuerman, R.W. Tear Analysis in Ocular Surface Diseases. Prog. Retin. Eye Res. 2012, 31, 527–550. [Google Scholar] [CrossRef] [PubMed]
  86. Pieragostino, D.; D’Alessandro, M.; di Ioia, M.; Di Ilio, C.; Sacchetta, P.; Del Boccio, P. Unraveling the Molecular Repertoire of Tears as a Source of Biomarkers: Beyond Ocular Diseases. Proteom. Clin. Appl. 2015, 9, 169–186. [Google Scholar] [CrossRef] [PubMed]
  87. Lebrecht, A.; Boehm, D.; Schmidt, M.; Koelbl, H.; Grus, F.H. Surface-Enhanced Laser Desorption/Ionisation Time-of-Flight Mass Spectrometry to Detect Breast Cancer Markers in Tears and Serum. Cancer Genom. Proteom. 2009, 6, 75–84. [Google Scholar]
  88. Morimoto, Y.; Conroy, S.M.; Franke, A.A.; Maskarinec, G. Nipple Aspirate Fluid Producer Status among Premenopausal Women in Hawaii. Breast J. 2012, 18, 504–505. [Google Scholar] [CrossRef]
  89. Suijkerbuijk, K.P.M.; Van Der Wall, E.; Meijrink, H.; Pan, X.; Rinkes, I.H.M.B.; Ausems, M.G.E.M.; Van Diest, P.J. Successful Oxytocin-Assisted Nipple Aspiration in Women at Increased Risk for Breast Cancer. Fam. Cancer 2010, 9, 321–325. [Google Scholar] [CrossRef]
  90. Shaheed, S.U.; Tait, C.; Kyriacou, K.; Mullarkey, J.; Burrill, W.; Patterson, L.H.; Linforth, R.; Salhab, M.; Sutton, C.W. Nipple Aspirate Fluid—A Liquid Biopsy for Diagnosing Breast Health. Proteom. Clin. Appl. 2017, 11, 1700015. [Google Scholar] [CrossRef]
  91. Shaheed, S.U.; Tait, C.; Kyriacou, K.; Linforth, R.; Salhab, M.; Sutton, C. Evaluation of Nipple Aspirate Fluid as a Diagnostic Tool for Early Detection of Breast Cancer. Clin. Proteom. 2018, 15, 3. [Google Scholar] [CrossRef]
  92. Patuleia, S.I.S.; Suijkerbuijk, K.P.M.; van der Wall, E.; van Diest, P.J.; Moelans, C.B. Nipple Aspirate Fluid at a Glance. Cancers 2022, 14, 159. [Google Scholar] [CrossRef]
  93. Chan, A.A.; Bashir, M.; Rivas, M.N.; Duvall, K.; Sieling, P.A.; Pieber, T.R.; Vaishampayan, P.A.; Love, S.M.; Lee, D.J. Characterization of the Microbiome of Nipple Aspirate Fluid of Breast Cancer Survivors. Sci. Rep. 2016, 6, 28061. [Google Scholar] [CrossRef] [PubMed]
  94. Bel’Skaya, L.V.; Sarf, E.A.; Solomatin, D.V.; Kosenok, V.K. Metabolic Features of Saliva in Breast Cancer Patients. Metabolites 2022, 12, 166. [Google Scholar] [CrossRef]
  95. Zhang, L.; Xiao, H.; Karlan, S.; Zhou, H.; Gross, J.; Elashoff, D.; Akin, D.; Yan, X.; Chia, D.; Karlan, B.; et al. Discovery and Preclinical Validation of Salivary Transcriptomic and Proteomic Biomarkers for the Non- Invasive Detection of Breast Cancer. PLoS ONE 2010, 5, e15573. [Google Scholar] [CrossRef] [PubMed]
  96. Cui, Y.; Yang, M.; Zhu, J.; Zhang, H.; Duan, Z.; Wang, S.; Liao, Z.; Liu, W. Developments in Diagnostic Applications of Saliva in Human Organ Diseases. Med. Nov. Technol. Devices 2022, 13, 100115. [Google Scholar] [CrossRef]
  97. Assad, D.X.; Acevedo, A.C.; Mascarenhas, E.C.P.; Normando, A.G.C.; Pichon, V.; Chardin, H.; Guerra, E.N.S.; Combes, A. Using an Untargeted Metabolomics Approach to Identify Salivary Metabolites in Women with Breast Cancer. Metabolites 2020, 10, 506. [Google Scholar] [CrossRef] [PubMed]
  98. Liu, S.Y.; Liao, Y.; Hosseinifard, H.; Imani, S.; Wen, Q.L. Diagnostic Role of Extracellular Vesicles in Cancer: A Comprehensive Systematic Review and Meta-Analysis. Front. Cell Dev. Biol. 2021, 9, 2749. [Google Scholar] [CrossRef] [PubMed]
  99. Lane, R.E.; Korbie, D.; Hill, M.M.; Trau, M. Extracellular Vesicles as Circulating Cancer Biomarkers: Opportunities and Challenges. Clin. Transl. Med. 2018, 7, 14. [Google Scholar] [CrossRef]
  100. Grölz, D.; Hauch, S.; Schlumpberger, M.; Guenther, K.; Voss, T.; Sprenger-Haussels, M.; Oelmüller, U. Liquid Biopsy Preservation Solutions for Standardized Pre-Analytical Workflows—Venous Whole Blood and Plasma. Curr. Pathobiol. Rep. 2018, 6, 275–286. [Google Scholar] [CrossRef]
  101. Salvianti, F.; Gelmini, S.; Costanza, F.; Mancini, I.; Sonnati, G.; Simi, L.; Pazzagli, M.; Pinzani, P. The Pre-Analytical Phase of the Liquid Biopsy. N. Biotechnol. 2020, 55, 19–29. [Google Scholar] [CrossRef]
  102. Lacroix, R.; Judicone, C.; Poncelet, P.; Robert, S.; Arnaud, L.; Sampol, J.; Dignat-George, F. Impact of Pre-Analytical Parameters on the Measurement of Circulating Microparticles: Towards Standardization of Protocol. J. Thromb. Haemost. 2012, 10, 437–446. [Google Scholar] [CrossRef]
  103. Abramowicz, A.; Widlak, P.; Pietrowska, M. Proteomic Analysis of Exosomal Cargo: The Challenge of High Purity Vesicle Isolation. Mol. Biosyst. 2016, 12, 1407–1419. [Google Scholar] [CrossRef]
  104. Van Deun, J.; Mestdagh, P.; Agostinis, P.; Akay, Ö.; Anand, S.; Anckaert, J.; Martinez, Z.A.; Baetens, T.; Beghein, E.; Bertier, L.; et al. EV-TRACK: Transparent Reporting and Centralizing Knowledge in Extracellular Vesicle Research. Nat. Methods 2017, 14, 228–232. [Google Scholar]
  105. Siwaponanan, P.; Keawvichit, R.; Lekmanee, K.; Chomanee, N.; Pattanapanyasat, K. Enumeration and Phenotyping of Circulating Microvesicles by Flow Cytometry and Nanoparticle Tracking Analysis: Plasma versus Serum. Int. J. Lab. Hematol. 2021, 43, 506–514. [Google Scholar] [CrossRef] [PubMed]
  106. Palviainen, M.; Saraswat, M.; Varga, Z.; Kitka, D.; Neuvonen, M.; Puhka, M.; Joenväärä, S.; Renkonen, R.; Nieuwland, R.; Takatalo, M.; et al. Extracellular Vesicles from Human Plasma and Serum Are Carriers of Extravesicular Cargo—Implications for Biomarker Discovery. PLoS ONE 2020, 15, e0236439. [Google Scholar] [CrossRef] [PubMed]
  107. Yekula, A.; Muralidharan, K.; Kang, K.M.; Wang, L.; Balaj, L.; Carter, B.S. From Laboratory to Clinic: Translation of Extracellular Vesicle Based Cancer Biomarkers. Methods 2020, 177, 58–66. [Google Scholar] [CrossRef]
  108. Daly, R.; O’Driscoll, L. Extracellular Vesicles in Blood: Are They Viable as Diagnostic and Predictive Tools in Breast Cancer? Drug Discov. Today 2021, 26, 778–785. [Google Scholar] [CrossRef] [PubMed]
  109. Hassis, M.E.; Niles, R.K.; Braten, M.N.; Albertolle, M.E.; Ewa Witkowska, H.; Hubel, C.A.; Fisher, S.J.; Williams, K.E. Evaluating the Effects of Pre-analytical Variables on the Stability of the Human Plasma Proteome. Anal. Biochem. 2015, 478, 14–22. [Google Scholar] [CrossRef] [PubMed]
  110. Bowen, R.A.R.; Adcock, D.M. Blood Collection Tubes as Medical Devices: The Potential to Affect Assays and Proposed Verification and Validation Processes for the Clinical Laboratory. Clin. Biochem. 2016, 49, 1321–1330. [Google Scholar] [CrossRef]
  111. Greco, V.; Piras, C.; Pieroni, L.; Urbani, A. Direct Assessment of Plasma/Serum Sample Quality for Proteomics Biomarker Investigation. Methods Mol. Biol. 2017, 1619, 3–21. [Google Scholar] [PubMed]
  112. Villanueva, J.; Philip, J.; Chaparro, C.A.; Li, Y.; Toledo-Crow, R.; DeNoyer, L.; Fleisher, M.; Robbins, R.J.; Tempst, P. Correcting Common Errors in Identifying Cancer-Specific Serum Peptide Signatures. J. Proteome Res. 2005, 4, 1060–1072. [Google Scholar] [CrossRef] [PubMed]
  113. Hsieh, S.Y.; Chen, R.K.; Pan, Y.H.; Lee, H.L. Systematical Evaluation of the Effects of Sample Collection Procedures on Low-Molecular-Weight Serum/Plasma Proteome Profiling. Proteomics 2006, 6, 3189–3198. [Google Scholar] [CrossRef] [PubMed]
  114. Bowen, R.A.R.; Hortin, G.L.; Csako, G.; Otañez, O.H.; Remaley, A.T. Impact of Blood Collection Devices on Clinical Chemistry Assays. Clin. Biochem. 2010, 43, 4–25. [Google Scholar] [CrossRef] [PubMed]
  115. Yin, P.; Peter, A.; Franken, H.; Zhao, X.; Neukamm, S.S.; Rosenbaum, L.; Lucio, M.; Zell, A.; Häring, H.U.; Xu, G.; et al. Pre-analytical Aspects and Sample Quality Assessment in Metabolomics Studies of Human Blood. Clin Chem 2013, 59, 833–845. [Google Scholar] [CrossRef]
  116. Barri, T.; Dragsted, L.O. UPLC-ESI-QTOF/MS and Multivariate Data Analysis for Blood Plasma and Serum Metabolomics: Effect of Experimental Artefacts and Anticoagulant. Anal. Chim. Acta 2013, 768, 118–128. [Google Scholar] [CrossRef]
  117. Dunn, W.B.; Broadhurst, D.; Begley, P.; Zelena, E.; Francis-Mcintyre, S.; Anderson, N.; Brown, M.; Knowles, J.D.; Halsall, A.; Haselden, J.N.; et al. Procedures for Large-Scale Metabolic Profiling of Serum and Plasma Using Gas Chromatography and Liquid Chromatography Coupled to Mass Spectrometry. Nat. Protoc. 2011, 6, 1060–1083. [Google Scholar] [CrossRef]
  118. Kamlage, B.; Maldonado, S.G.; Bethan, B.; Peter, E.; Schmitz, O.; Liebenberg, V.; Schatz, P. Quality Markers Addressing Pre-analytical Variations of Blood and Plasma Processing Identified by Broad and Targeted Metabolite Profiling. Clin. Chem. 2014, 60, 399–412. [Google Scholar] [CrossRef]
  119. Liu, X.; Hoene, M.; Yin, P.; Fritsche, L.; Plomgaard, P.; Hansen, J.S.; Nakas, C.T.; Niess, A.M.; Hudemann, J.; Haap, M.; et al. Quality Control of Serum and Plasma by Quantification of (4E,14Z)-Sphingadienine-C18-1-Phosphate Uncovers Common Pre-analytical Errors during Handling of Whole Blood. Clin. Chem. 2018, 64, 810–819. [Google Scholar] [CrossRef]
  120. Teahan, O.; Gamble, S.; Holmes, E.; Waxman, J.; Nicholson, J.K.; Bevan, C.; Keun, H.C. Impact of Analytical Bias in Metabonomic Studies of Human Blood Serum and Plasma. Anal. Chem. 2006, 78, 4307–4318. [Google Scholar] [CrossRef]
  121. Halvey, P.; Farutin, V.; Koppes, L.; Gunay, N.S.; Pappas, D.A.; Manning, A.M.; Capila, I. Variable Blood Processing Procedures Contribute to Plasma Proteomic Variability. Clin. Proteom. 2021, 18, 5. [Google Scholar] [CrossRef]
  122. Lippi, G.; Blanckaert, N.; Bonini, P.; Green, S.; Kitchen, S.; Palicka, V.; Vassault, A.J.; Plebani, M. Haemolysis: An Overview of the Leading Cause of Unsuitable Specimens in Clinical Laboratories. Clin. Chem. Lab. Med. 2008, 46, 764–772. [Google Scholar] [CrossRef] [PubMed]
  123. Ammerlaan, W.; Trezzi, J.P.; Lescuyer, P.; Mathay, C.; Hiller, K.; Betsou, F. Method Validation for Preparing Serum and Plasma Samples from Human Blood for Downstream Proteomic, Metabolomic, and Circulating Nucleic Acid-Based Applications. Biopreserv. Biobank. 2014, 12, 269–280. [Google Scholar] [CrossRef]
  124. Stevens, V.L.; Hoover, E.; Wang, Y.; Zanetti, K.A. Pre-Analytical Factors That Affect Metabolite Stability in Human Urine, Plasma, and Serum: A Review. Metabolites 2019, 9, 156. [Google Scholar] [CrossRef] [PubMed]
  125. Züllig, T.; Trötzmüller, M.; Köfeler, H.C. Lipidomics from Sample Preparation to Data Analysis: A Primer. Anal Bioanal Chem 2020, 412, 2191–2209. [Google Scholar] [CrossRef]
  126. Scherer, M.; Schmitz, G.; Liebisch, G. High-Throughput Analysis of Sphingosine 1-Phosphate, Sphinganine 1-Phosphate, and Lysophosphatidic Acid in Plasma Samples by Liquid Chromatography—Tandem Mass Spectrometry. Clin. Chem. 2009, 55, 1218–1222. [Google Scholar] [CrossRef]
  127. Jain, M.; Kennedy, A.D.; Elsea, S.H.; Miller, M.J. Analytes Related to Erythrocyte Metabolism Are Reliable Biomarkers for Pre-analytical Error Due to Delayed Plasma Processing in Metabolomics Studies. Clin. Chim. Acta 2017, 466, 105–111. [Google Scholar] [CrossRef] [PubMed]
  128. Barelli, S.; Crettaz, D.; Thadikkaran, L.; Rubin, O.; Tissot, J.D. Plasma/Serum Proteomics: Pre-Analytical Issues. Expert Rev. Proteom. 2007, 4, 363–370. [Google Scholar] [CrossRef]
  129. Lesche, D.; Geyer, R.; Lienhard, D.; Nakas, C.T.; Diserens, G.; Vermathen, P.; Leichtle, A.B. Does Centrifugation Matter? Centrifugal Force and Spinning Time Alter the Plasma Metabolome. Metabolomics 2016, 12, 159. [Google Scholar] [CrossRef]
  130. Rai, A.J.; Vitzthum, F. Effects of Pre-analytical Variables on Peptide and Protein Measurements in Human Serum and Plasma: Implications for Clinical Proteomics. Expert Rev. Proteom. 2006, 3, 409–426. [Google Scholar] [CrossRef]
  131. Valo, E.; Colombo, M.; Sandholm, N.; McGurnaghan, S.J.; Blackbourn, L.A.K.; Dunger, D.B.; McKeigue, P.M.; Forsblom, C.; Groop, P.H.; Colhoun, H.M.; et al. Effect of Serum Sample Storage Temperature on Metabolomic and Proteomic Biomarkers. Sci. Rep. 2022, 12, 4571. [Google Scholar] [CrossRef]
  132. Ferguson, R.E.; Hochstrasser, D.F.; Banks, R.E. Impact of Pre-analytical Variables on the Analysis of Biological Fluids in Proteomic Studies. Proteom. Clin. Appl. 2007, 1, 739–746. [Google Scholar] [CrossRef] [PubMed]
  133. Zander, J.; Bruegel, M.; Kleinhempel, A.; Becker, S.; Petros, S.; Kortz, L.; Dorow, J.; Kratzsch, J.; Baber, R.; Ceglarek, U.; et al. Effect of Biobanking Conditions on Short-Term Stability of Biomarkers in Human Serum and Plasma. Clin. Chem. Lab. Med. 2014, 52, 629–639. [Google Scholar] [CrossRef] [PubMed]
  134. Lee, D.H.; Kim, J.W.; Jeon, S.Y.; Park, B.K.; Han, B.G. Proteomic Analysis of the Effect of Storage Temperature on Human Serum. Ann. Clin. Lab. Sci. 2010, 40, 61–70. [Google Scholar] [PubMed]
  135. Mitchell, B.L.; Yasui, Y.; Li, C.I.; Fitzpatrick, A.L.; Lampe, P.D. Impact of Freeze-Thaw Cycles and Storage Time on Plasma Samples Used in Mass Spectrometry Based Biomarker Discovery Projects. Cancer Inform. 2005, 1, 98–104. [Google Scholar] [CrossRef]
  136. Fliniaux, O.; Gaillard, G.; Lion, A.; Cailleu, D.; Mesnard, F.; Betsou, F. Influence of Common Pre-analytical Variations on the Metabolic Profile of Serum Samples in Biobanks. J. Biomol. NMR 2011, 51, 457–465. [Google Scholar] [CrossRef]
  137. Gardner, A.; Carpenter, G.; So, P.W. Salivary Metabolomics: From Diagnostic Biomarker Discovery to Investigating Biological Function. Metabolites 2020, 10, 47. [Google Scholar] [CrossRef]
  138. Li, Y.; Xun, D.; Li, L.; Wang, B.; Lv, J.; Liu, H.; Zhu, L.; Ma, F.; Chen, X.; Tian, S.; et al. Deep Dive on the Proteome of Human Body Fluids: A Valuable Data Resource for Biomarker Discovery. Cancer Genom. Proteom. 2021, 18, 549–568. [Google Scholar] [CrossRef]
  139. Dayon, L.; Cominetti, O.; Affolter, M. Proteomics of Human Biological Fluids for Biomarker Discoveries: Technical Advances and Recent Applications. Expert Rev. Proteom. 2022, 19, 131–151. [Google Scholar] [CrossRef]
  140. Panneerselvam, K.; Ishikawa, S.; Krishnan, R.; Sugimoto, M. Salivary Metabolomics for Oral Cancer Detection: A Narrative Review. Metabolites 2022, 12, 436. [Google Scholar] [CrossRef]
  141. Srivastava, A.; Creek, D.J. Discovery and Validation of Clinical Biomarkers of Cancer: A Review Combining Metabolomics and Proteomics. Proteomics 2019, 19, e1700448. [Google Scholar] [CrossRef]
  142. Pusch, W.; Kostrzewa, M. Application of MALDI-TOF Mass Spectrometry in Screening and Diagnostic Research. Curr. Pharm. Des. 2005, 11, 2577–2591. [Google Scholar] [CrossRef] [PubMed]
  143. Hosnedlova, B.; Kepinska, M.; Ruttkay-Nedecky, B.; Fernandez, C.; Parak, T.; Milnerowicz, H.; Sochor, J.; Bjørklund, G.; Kizek, R. Matrix Assisted Laser Desorption/Ionization as a New Cancer Diagnostic Tool. In Encyclopedia of Biomedical Engineering; Elsevier: Cambridge, MA, USA, 2019; Volume 1–3, pp. 400–414. ISBN 9780128051443. [Google Scholar]
  144. Gutierrez, J.A.; Dorocke, J.A.; Knierman, M.D.; Gelfanova, V.; Higgs, R.E.; Koh, N.L.; Hale, J.E. Quantitative Determination of Peptides Using Matrix-Assisted Laser Desorption/Ionization Time-of-Flight Mass Spectrometry. Biotechniques 2005, 38, S13–S17. [Google Scholar] [CrossRef] [PubMed]
  145. Albrethsen, J. Reproducibility in Protein Profiling by MALDI-TOF Mass Spectrometry. Clin. Chem. 2007, 53, 852–858. [Google Scholar] [CrossRef] [PubMed]
  146. Kiehntopf, M.; Siegmund, R.; Deufel, T. Use of SELDI-TOF Mass Spectrometry for Identification of New Biomarkers: Potential and Limitations. Clin. Chem. Lab. Med. 2007, 45, 1435–1449. [Google Scholar] [CrossRef]
  147. Hu, T.; Zhang, J.L. Mass-Spectrometry-Based Lipidomics. J. Sep. Sci. 2018, 41, 351–372. [Google Scholar] [CrossRef]
  148. Xie, F.; Liu, T.; Qian, W.J.; Petyuk, V.A.; Smith, R.D. Liquid Chromatography-Mass Spectrometry-Based Quantitative Proteomics. J. Biol. Chem. 2011, 286, 25443–25449. [Google Scholar] [CrossRef]
  149. Grebe, S.K.G.; Singh, R.J. LC-MS/MS in the Clinical Laboratory—Where to from Here? Clin. Biochem. Rev. 2011, 32, 5–31. [Google Scholar]
  150. Lubes, G.; Goodarzi, M. GC–MS Based Metabolomics Used for the Identification of Cancer Volatile Organic Compounds as Biomarkers. J. Pharm. Biomed. Anal. 2018, 147, 313–322. [Google Scholar] [CrossRef]
  151. Emwas, A.H.M. The Strengths and Weaknesses of NMR Spectroscopy and Mass Spectrometry with Particular Focus on Metabolomics Research. Methods Mol. Biol. 2015, 1277, 161–193. [Google Scholar] [CrossRef]
  152. Meftahi, G.H.; Bahari, Z.; Zarei Mahmoudabadi, A.; Iman, M.; Jangravi, Z. Applications of Western Blot Technique: From Bench to Bedside. Biochem. Mol. Biol. Educ. 2021, 49, 509–517. [Google Scholar] [CrossRef]
  153. Monteoliva, L.; Albar, J.P. Differential Proteomics: An Overview of Gel and Non-Gel Based Approaches. Brief. Funct. Genom. Proteom. 2004, 3, 220–239. [Google Scholar] [CrossRef] [PubMed]
  154. Baggerman, G.; Vierstraete, E.; De Loof, A.; Schoofs, L. Gel-Based Versus Gel-Free Proteomics: A Review. Comb. Chem. High Throughput Screen. 2005, 8, 669–677. [Google Scholar] [CrossRef] [PubMed]
  155. Meleady, P. Two-Dimensional Gel Electrophoresis and 2D-DIGE. Methods Mol. Biol. 2018, 1664, 3–14. [Google Scholar] [PubMed]
  156. Beckett, P. The Basics of 2D DIGE. Methods Mol. Biol. 2012, 854, 9–19. [Google Scholar] [CrossRef]
  157. Hosseini, S.; Vázquez-Villegas, P.; Rito-Palomares, M.; Martinez-Chapa, S.O. Advantages, Disadvantages and Modifications of Conventional ELISA. Springer Briefs in Applied Sciences and Technology; Springer: Singapore, 2018; pp. 67–115. [Google Scholar]
  158. Sakamoto, S.; Putalun, W.; Vimolmangkang, S.; Phoolcharoen, W.; Shoyama, Y.; Tanaka, H.; Morimoto, S. Enzyme-Linked Immunosorbent Assay for the Quantitative/Qualitative Analysis of Plant Secondary Metabolites. J. Nat. Med. 2018, 72, 32–42. [Google Scholar] [CrossRef]
  159. Wang, K.; Huang, C.; Nice, E.C. Proteomics, Genomics and Transcriptomics: Their Emerging Roles in the Discovery and Validation of Colorectal Cancer Biomarkers. Expert Rev. Proteom. 2014, 11, 179–205. [Google Scholar] [CrossRef]
  160. Bratulic, S.; Gatto, F.; Nielsen, J. The Translational Status of Cancer Liquid Biopsies. Regen. Eng. Transl. Med. 2021, 7, 312–352. [Google Scholar] [CrossRef]
  161. Hajduk, J.; Matysiak, J.; Kokot, Z.J. Challenges in Biomarker Discovery with MALDI-TOF MS. Clin. Chim. Acta 2016, 458, 84–98. [Google Scholar] [CrossRef]
  162. Chandramouli, K.; Qian, P.-Y. Proteomics: Challenges, Techniques and Possibilities to Overcome Biological Sample Complexity. Hum. Genom. Proteom. 2009, 1, 239204. [Google Scholar] [CrossRef]
  163. Jiang, M.; Gu, G.; Ni, B.; Wang, W.; Shi, J.; Liao, P.; Hu, H. Detection of Serum Protein Biomarkers by Surface Enhanced Laser Desorption/Ionization in Patients with Adenocarcinoma of the Lung. Asia Pac. J. Clin. Oncol. 2014, 10, e7–e12. [Google Scholar] [CrossRef]
  164. Muthu, M.; Vimala, A.; Mendoza, O.H.; Gopal, J. Tracing the Voyage of SELDI-TOF MS in Cancer Biomarker Discovery and Its Current Depreciation Trend—Need for Resurrection? TrAC Trends Anal. Chem. 2016, 76, 95–101. [Google Scholar] [CrossRef]
  165. Van, Q.N.; Veenstra, T.D. How Close Is the Bench to the Bedside? Metabolic Profiling in Cancer Research. Genome Med. 2009, 1, 5. [Google Scholar] [CrossRef] [PubMed]
  166. Luo, C.; Zhang, X.; He, Y.; Chen, H.; Liu, M.; Wang, H.; Tang, L.; Tu, G.; Ding, M. A Pseudo-Targeted Metabolomics Study Based on Serum Bile Acids Profiling for the Differential Diagnosis of Benign and Malignant Breast Lesions. Steroids 2021, 175, 108914. [Google Scholar] [CrossRef] [PubMed]
  167. Marian, C.; Varghese, R.S.; Ahn, J.; Da Cunha, P.A.; Willey, S.; Sidawy, M.; Rone, J.D.; Cheema, A.K.; Luta, G.; Nezami Ranjbar, M.R.; et al. Metabolomic Profiling of Breast Tumors Using Ductal Fluid. Int. J. Oncol. 2016, 49, 2245–2254. [Google Scholar] [CrossRef]
  168. Oktay, K.; Santaliz-Casiano, A.; Patel, M.; Marino, N.; Storniolo, A.M.V.; Torun, H.; Acar, B.; Madak Erdogan, Z. A Computational Statistics Approach to Evaluate Blood Biomarkers for Breast Cancer Risk Stratification. Horm. Cancer 2020, 11, 17–33. [Google Scholar] [CrossRef]
  169. Hadi, N.I.; Jamal, Q.; Iqbal, A.; Shaikh, F.; Somroo, S.; Musharraf, S.G. Serum Metabolomic Profiles for Breast Cancer Diagnosis, Grading and Staging by Gas Chromatography-Mass Spectrometry. Sci. Rep. 2017, 7, 1715. [Google Scholar] [CrossRef]
  170. Rashed, R.; Darwish, H.; Omran, M.; Belal, A.; Zahran, F. A Novel Serum Metabolome Score for Breast Cancer Diagnosis. Br. J. Biomed. Sci. 2020, 77, 196–201. [Google Scholar] [CrossRef]
  171. Jové, M.; Collado, R.; Quiles, J.L.; Ramírez-Tortosa, M.C.; Sol, J.; Ruiz-Sanjuan, M.; Fernandez, M.; Cabrera, C.d.l.T.; Ramírez-Tortosa, C.; Granados-Principal, S.; et al. A Plasma Metabolomic Signature Discloses Human Breast Cancer. Oncotarget 2017, 8, 19522–19533. [Google Scholar] [CrossRef]
  172. Huang, J.; Sun, J.; Chen, Y.; Song, Y.; Dong, L.; Zhan, Q.; Zhang, R.; Abliz, Z. Analysis of Multiplex Endogenous Estrogen Metabolites in Human Urine Using Ultra-Fast Liquid Chromatography-Tandem Mass Spectrometry: A Case Study for Breast Cancer. Anal. Chim. Acta 2012, 711, 60–68. [Google Scholar] [CrossRef]
  173. Hadi, N.I.; Jamal, Q. “OMIC” Tumor Markers for Breast Cancer: A Review. Pak. J. Med. Sci. 2015, 31, 1256. [Google Scholar] [CrossRef]
  174. Huang, Q.; Tan, Y.; Yin, P.; Ye, G.; Gao, P.; Lu, X.; Wang, H.; Xu, G. Metabolic Characterization of Hepatocellular Carcinoma Using Nontargeted Tissue Metabolomics. Cancer Res. 2013, 73, 4992–5002. [Google Scholar] [CrossRef] [PubMed]
  175. Chen, T.; Xie, G.; Wang, X.; Fan, J.; Qiu, Y.; Zheng, X.; Qi, X.; Cao, Y.; Su, M.; Wang, X.; et al. Serum and Urine Metabolite Profiling Reveals Potential Biomarkers of Human Hepatocellular Carcinoma. Mol. Cell. Proteom. 2011, 10. [Google Scholar] [CrossRef]
  176. Ghosh, A.; Nishtala, K. Biofluid Lipidome: A Source for Potential Diagnostic Biomarkers. Clin. Transl. Med. 2017, 6, 22. [Google Scholar] [CrossRef] [PubMed]
  177. Yang, K.; Han, X. Lipidomics: Techniques, Applications, and Outcomes Related to Biomedical Sciences. Trends Biochem. Sci. 2016, 41, 954–969. [Google Scholar] [CrossRef] [PubMed]
  178. Wang, J.; Wang, C.; Han, X. Tutorial on Lipidomics. Anal. Chim. Acta 2019, 1061, 28–41. [Google Scholar] [CrossRef] [PubMed]
  179. Li, J.; Vosegaard, T.; Guo, Z. Applications of Nuclear Magnetic Resonance in Lipid Analyses: An Emerging Powerful Tool for Lipidomics Studies. Prog. Lipid Res. 2017, 68, 37–56. [Google Scholar] [CrossRef]
  180. Zhang, F.; Ge, W.; Ruan, G.; Cai, X.; Guo, T. Data-Independent Acquisition Mass Spectrometry-Based Proteomics and Software Tools: A Glimpse in 2020. Proteomics 2020, 20, e1900276. [Google Scholar] [CrossRef]
  181. Ràfols, P.; Vilalta, D.; Brezmes, J.; Cañellas, N.; del Castillo, E.; Yanes, O.; Ramírez, N.; Correig, X. Signal Preprocessing, Multivariate Analysis and Software Tools for MA(LDI)-TOF Mass Spectrometry Imaging for Biological Applications. Mass Spectrom. Rev. 2018, 37, 281–306. [Google Scholar] [CrossRef]
  182. Teleman, J.; Röst, H.L.; Rosenberger, G.; Schmitt, U.; Malmström, L.; Malmström, J.; Levander, F. DIANA-Algorithmic Improvements for Analysis of Data-Independent Acquisition MS Data. Bioinformatics 2015, 31, 555–562. [Google Scholar] [CrossRef]
  183. Mertens, B.J.A. Transformation, Normalization, and Batch Effect in the Analysis of Mass Spectrometry Data for Omics Studies. In Statistical Analysis of Proteomics, Metabolomics, and Lipidomics Data Using Mass Spectrometry; Springer: Cham, Switzerland, 2017; pp. 1–21. [Google Scholar]
  184. Välikangas, T.; Suomi, T.; Elo, L.L. A Systematic Evaluation of Normalization Methods in Quantitative Label-Free Proteomics. Brief. Bioinform. 2018, 19, 1–11. [Google Scholar] [CrossRef]
  185. Vafaee, F.; Diakos, C.; Kirschner, M.B.; Reid, G.; Michael, M.Z.; Horvath, L.G.; Alinejad-Rokny, H.; Cheng, Z.J.; Kuncic, Z.; Clarke, S. A Data-Driven, Knowledge-Based Approach to Biomarker Discovery: Application to Circulating MicroRNA Markers of Colorectal Cancer Prognosis. NPJ Syst. Biol. Appl. 2018, 4, 20. [Google Scholar] [CrossRef] [PubMed]
  186. Vijayan, A.; Fatima, S.; Sowmya, A.; Vafaee, F. Blood-Based Transcriptomic Signature Panel Identification for Cancer Diagnosis: Benchmarking of Feature Extraction Methods. Brief. Bioinform. 2022, 23, bbac315. [Google Scholar] [CrossRef]
  187. Liu, L.; Chen, X.; Petinrin, O.O.; Zhang, W.; Rahaman, S.; Tang, Z.R.; Wong, K.C. Machine Learning Protocols in Early Cancer Detection Based on Liquid Biopsy: A Survey. Life 2021, 11, 638. [Google Scholar] [CrossRef] [PubMed]
  188. Zhang, X.; Jonassen, I.; Goksøyr, A. Machine Learning Approaches for Biomarker Discovery Using Gene Expression Data. In Bioinformatics; Exon Publications: Brisbane, QLD, Australia, 2021; pp. 53–64. [Google Scholar]
  189. Moons, K.G.M.; de Groot, J.A.H.; Bouwmeester, W.; Vergouwe, Y.; Mallett, S.; Altman, D.G.; Reitsma, J.B.; Collins, G.S. Critical Appraisal and Data Extraction for Systematic Reviews of Prediction Modelling Studies: The CHARMS Checklist. PLoS Med. 2014, 11, e1001744. [Google Scholar] [CrossRef] [PubMed]
  190. Darrow, J.J.; Avorn, J.; Kesselheim, A.S. FDA Regulation and Approval of Medical Devices: 1976–2020. JAMA J. Am. Med. Assoc. 2021, 326, 420–432. [Google Scholar] [CrossRef]
  191. Rathi, V.; Wright, G.; Constantin, D.; Chang, S.; Pham, H.; Jones, K.; Palios, A.; Mclachlan, S.A.; Conron, M.; McKelvie, P.; et al. Clinical Validation of the 50 Gene AmpliSeq Cancer Panel V2 for Use on a next Generation Sequencing Platform Using Formalin Fixed, Paraffin Embedded and Fine Needle Aspiration Tumour Specimens. Pathology 2017, 49, 75–82. [Google Scholar] [CrossRef]
  192. Wang, P.; Kricka, L.J. Current and Emerging Trends in Point-of-Care Technology and Strategies for Clinical Validation and Implementation. Clin. Chem. 2018, 64, 1439–1452. [Google Scholar] [CrossRef]
  193. Klein, E.A.; Richards, D.; Cohn, A.; Tummala, M.; Lapham, R.; Cosgrove, D.; Chung, G.; Clement, J.; Gao, J.; Hunkapiller, N.; et al. Clinical Validation of a Targeted Methylation-Based Multi-Cancer Early Detection Test Using an Independent Validation Set. Ann. Oncol. 2021, 32, 1167–1177. [Google Scholar] [CrossRef]
  194. Nadauld, L.D.; McDonnell, C.H.; Beer, T.M.; Liu, M.C.; Klein, E.A.; Hudnut, A.; Whittington, R.A.; Taylor, B.; Oxnard, G.R.; Lipson, J.; et al. The Pathfinder Study: Assessment of the Implementation of an Investigational Multi-Cancer Early Detection Test into Clinical Practice. Cancers 2021, 13, 3501. [Google Scholar] [CrossRef]
  195. Chen, G.; Zhang, J.; Fu, Q.; Taly, V.; Tan, F. Integrative Analysis of Multi-Omics Data for Liquid Biopsy. Br. J. Cancer 2023, 128, 505–518. [Google Scholar] [CrossRef]
Figure 1. Variable factors involved in biomarker discovery pipeline. This schematic flow chart illustrates the influential factors involved in typical pre-analytical, analytical, and post-analytical stages in proteomics, metabolomics, and lipidomic investigations in breast cancer liquid biopsy.
Figure 1. Variable factors involved in biomarker discovery pipeline. This schematic flow chart illustrates the influential factors involved in typical pre-analytical, analytical, and post-analytical stages in proteomics, metabolomics, and lipidomic investigations in breast cancer liquid biopsy.
Cancers 15 02780 g001
Figure 2. The proportion of metabolomic, lipidomic, and proteomic investigations carried out on various biofluid samples of breast cancer between January 2001 and April 2023. (A) Percentage of metabolomic, lipidomic, and proteomic studies according to their biofluid source. (B) The trend in studying non-nucleotide-based biomarkers from various biofluids of breast cancer in the last twenty years.
Figure 2. The proportion of metabolomic, lipidomic, and proteomic investigations carried out on various biofluid samples of breast cancer between January 2001 and April 2023. (A) Percentage of metabolomic, lipidomic, and proteomic studies according to their biofluid source. (B) The trend in studying non-nucleotide-based biomarkers from various biofluids of breast cancer in the last twenty years.
Cancers 15 02780 g002
Table 1. Protocol variability in breast cancer-associated biomarker discovery workflows.
Table 1. Protocol variability in breast cancer-associated biomarker discovery workflows.
AimPre-Analytical PhaseAnalytical PhasePost-Analytical PhaseRef
BioSourceCollection TubeTime to Sample ProcessingCentrifugationStorageTumour GradeTechniqueValidation MethodHypothesis Test Performed
ProteomicSerumNA4 °C for 1–2 h3000 rpm for 5 min + 12,000 rpm for 5 min−80 °CNASELDI-TOF-MSSDS-PAGE
MALDI-TOF/TOF
  • t-test
  • ANOVA
[26]
SerumPlastic tube with clot activator15 min3280× g for 5 min, 4 °C−80 °CNASELDI-TOF
MALDI-TOF-TOF
NA
  • t-tests
  • Multivariate discrimination analysis
  • ANOVA
  • ANN
  • ROC
[27]
PlasmaK2EDTA tube2 h1300× g for 10 min−80 °CNA1D gel electrophoresis
2D gel electrophoresis
LC-MS/MS
WBUnpaired t-test[28]
PlasmaEDTA tube30 min4000× g for 30 min−80 °CNALC-MS/MSWBt-test[29]
PlasmaSodium EDTA tubeNA1400× g for 5 min, 4 °CNDLow and high gradeLabel-free nano-LC/MSMSWBMann–Whitney[30]
NAFGraduated micropipetteImmediately1500 rpm for 10 min−80 °CI/IISELDI-TOF-MSELISASupervised and unsupervised cluster analysis[14]
NAFTube pre-treated
with cocktail mixture of protease inhibitor
<30 minNAST: −20 °C
LT: −80 °C
I–III1D LC-MS/MSNA
  • Pearson’s correlation coefficients
  • Paired Student t-test
[31]
UrineSterile tubeImmediately2000× g for 10 min, 4 °CST: −20 °C
LT: −80 °C
II–IIILabel-free LC-MS/MSWBANOVA[9]
First Morning UrineTube containing 0.02% w/v Sodium Azide)NANANDI/IIStandardisation phase: 2D gel electrophoresis
Discovery phase: 2D-DIGE, MALDI-TOF-TOF, SWATH-MS, iTRAQ, LC-QTOF
WB
MRM
  • Supervised and unsupervised cluster analysis
  • Multivariate analysis
  • Chi-square
[10]
MetabolomicPlasmaEDTA tube<2 h3000× g for 10 min, 4 °C−80 °CI–IIILC-MSNA
  • Kruskal–Wallis
  • Mann–Whitney U test
  • ROC
[32]
PlasmaK2EDTA tubeImmediately1500× g for 10 min, RT−80 °CI–IIILC-QTOF-MS
LC-QQQ-MS
NA
  • Student’s t-test
  • PLS-DA
  • OPLS-DA
[33]
SerumVacutainer tube30 min3000 rpm for 10 min, 4 °C−80 °CI–IIIUHPLC-QTOF-(ESIþ)-MSNA
  • Pearson
  • ROC
  • PCA
  • PLS-DA
  • t-test
[34]
First Morning UrineNANA3000× g for 10 min, RT−80 °CI/IIIGC–MS
LC-QTOF/MS
NA
  • PCA
  • OPLS-DA
  • Univariate analysis
  • Unpaired t-test
  • Mann–Whitney U test
[35]
SalivaPolypropylene tubeNANA−80 °C0–IVCE-TOF-MSLC-QQQ-MS
  • Mann–Whitney U test
  • Kruskal–Wallis
  • Multiple logistic regression
  • Multiple AD tree models
[36]
SalivaNA10 min13,500 rpm for 20 min, 4 °C−40 °CI–IVHILIC-ESI-MS
RPLC-ESI-MS
NA
  • Mann–Whitney U test
  • PLS-DA
  • PCA
[37]
LipidomicPlasmaHeparin tubeNA1500× g for 15 min−80 °CI/IIUPLC-QTOF/MSNA
  • t-test
  • One-way ANOVA
  • OPLS-D
[38]
PlasmaEDTA tube<2 h2600× g for 10 min, 4 °C−80 °C0- IILC-ESI-MS/MSNA
  • t-test
  • Binary logical regression
  • ROC
[39]
SerumNANANA−80 °CNANMR spectroscopyNA
  • t-test
  • Mann–Whitney U test
  • Chi-square
  • Binary logistic regression
[40]
First Morning UrineNANA3000× g for 10 min, RT−80 °CI/IIILC–MSNA
  • OPLS-DA
  • Univariate analysis
  • Unpaired t-test
  • Mann–Whitney U test
  • ROC
[35]
SalivaPolypropylene tubeNA10,000× g for 10 minWithout freezing and storageI–IIIIR spectroscopyNA
  • Mann–Whitney U-test
  • Kruskal–Wallis
  • Multivariate comparison
[41]
NA: Not Available, EDTA: Ethylenediaminetetraacetic Acid, K2 EDTA: Dipotassium Ethylenediaminetetraacetic Acid, h: Hour, min: Minute, RT: Room Temperature, ST: Short-Term, LT: Long-Term, WB: Western Blotting, ELISA: Enzyme-linked Immunosorbent Assay, ANN: Artificial Neural Network, ROC: Receiver Operating Characteristic.
Table 2. Pre-analytical variables during sample collection and handling.
Table 2. Pre-analytical variables during sample collection and handling.
Pre-Analytical VariableLiterature Findings
Collection Tubes
  • Sample collection containers are frequently overlooked variables in laboratory settings [110]
  • The same sample might have different protein profiles when collected in two different types of tube [111,112,113]
  • Blood tube components may adsorb some analytes, particularly proteins, leading to their detection loss [114]
  • Release of plasticisers from tubes into samples may adversely affect high-resolution mass spectrometric examinations [62]
Anti-Coagulant
  • For metabolomics profiling, sodium fluoride (NaF) and EDTA salts caused less interference than sodium citrate or lithium-heparin [115]
  • Heparin [116,117] or EDTA plasma [115,118,119] is recommended for mass spectrometry-based lipidomic and metabolomic analyses; EDTA plasma is unsuitable for NMR-based approaches as it leads to interferences in the spectra [120]
  • EDTA anti-coagulant is preferable for proteomics [111,121]
Hemolysis
  • One of the most common pre-analytical errors [122]
  • Destruction of red blood cells
  • Release of proteins, metabolites, and lipids into serum or plasma [115,123]
  • May obstruct correct profile interpretation [115,122]
  • MS-based assessments may be affected [124]
Incubation Time
  • Many chemical and enzymatic reactions will continue and eventually metabolise the lipids [125,126]
  • Blood cells constantly release, uptake, and metabolise compounds [69,119,127]
  • Metabolites are more sensitive to prolonged incubation at room temperature than at 0–4 °C [124]
  • Peptides and degraded proteins can be released from blood cells [128]
Centrifugation Force
  • Minor differences in centrifugation could lead to variations in metabolomic patterns [129]
  • Higher centrifugation (between 2300 and 4000× g for 5–10 min) is recommended for lipidomic and metabolomic studies [69]
  • Centrifugation at 1300–2000× g for 15 min was recommended for proteomic studies [130]
Storage Conditions
  • Several analytes can be affected by storage temperature and time [131]
  • Serum proteins change more at room temperature compared with −20 °C and −80 °C [132]
  • Storage at lower temperatures, such as −80 °C, is recommended [133,134]
Freeze–Thaw Cycles
  • Repeated freeze–thaw of samples can result in profile alterations [135,136]
  • One freeze–thaw cycle leads to dramatic alterations in several urinary proteins [132]
Table 3. Advantages and disadvantages of various analytical techniques used for proteins, metabolites, and lipidomics biomarkers.
Table 3. Advantages and disadvantages of various analytical techniques used for proteins, metabolites, and lipidomics biomarkers.
TechniquesAdvantagesLimitationsBiomarker Type
MALDI-TOF-MS
[142,143,144,145]
  • Rapid and straightforward operability
  • Low sample volumes
  • Mostly single-charged registered ions [M-H]+
  • High throughput
  • High accuracy, resolution, and sensitivity
  • No staining, labelling, anti-body, and hybridisation
  • Suitable for large polypeptides (>30 kDa) detection
  • Variation in the surface of the MALDI-TOF target
  • Limited dynamic range
  • Sensitive to contaminants
  • Low reproducibility
  • Proteins
SELDI-TOF-MS
[146]
  • High throughput
  • Low sample volumes
  • High sensitivity
  • Easy operability
  • Suitable for small peptides (∼500 Da) detection
  • Suitable for low MW, modified, truncated, or fragmented proteins detection
  • Failure of the validation process
  • Low reproducibility
  • Low resolution
  • Biased toward smaller peptides and proteins (<30 kDa)
  • Problems in larger MW proteins and PTM identification
  • Ion suppression
  • Prone to artefacts generation
  • Proteins
LC-MS
[68,147,148,149,150]
  • High throughput
  • High resolution
  • Suitable for low and high-molecular-weight compounds
  • High sensitivity
  • Problems in identifying hydrocarbons that produce similar ions
  • Highly manual workflows for sample preparation can benefit from automation
  • The high complexity of the instrumentation’s operation and maintenance when looking at a limited number of analytes
  • Proteins
  • Metabolites
  • Lipids
GC-MS
[147,150]
  • High-efficiency separations
  • Suitable for nonpolar, volatile, and small molecules
  • High sensitivity
  • High throughput
  • Limited mass range
  • Limited to thermally stable and volatile compounds
  • Destructive analysis
  • Not suitable for compounds heavier than 1000 Da
  • Time-consuming for sample preparation
  • Metabolites
  • Lipids
NMR
[150,151]
  • Very high reproducibility
  • High throughput
  • Non-destructive Sample recovery
  • Rapid
  • Highly skilled operators
  • Low sensitivity
  • Cost is higher than GC-MS and LC-MS
  • Difficult to quantify the noise.
  • Metabolites
1DGE
[68,152,153,154]
  • Simple workflow
  • Rapid
  • Cost-effective
  • Limited reproducibility
  • Unsuitable for low-abundance proteins
  • Hydrophobic proteins’ insolubility
  • Proteins
2DGE
[68,152,153,154]
  • High resolution
  • High throughput
  • Cost-effective
  • Gel-to-gel variation
  • Lack of sensitivity
  • Poor dynamic range
  • Time-consuming
  • Highly skilled operators
  • Not automated approach
  • Proteins
2D-DIGE
[153,155,156]
  • Wide dynamic range detection
  • Fewer number of gels
  • Straightforward matching between gels
  • Higher sensitivity and reproducibility over 2DGE
  • Highly skilled operators
  • Time-consuming
  • Lower throughput
  • Not suitable for extremely acidic, basic, or hydrophobic proteins
  • Proteins
Immunoassay techniques
(ELISA, Western Blot)
[152,157,158]
  • High sensitivity and specificity when looking at a limited number of analytes
  • Cost-effective
  • Simple workflows
  • Highly reproducible
  • Suitable for validation
  • Resource-intensive efforts
  • Time-consuming
  • Not recognition of posttranslational protein variants
  • Limited multiplexing options
  • Relatively high sample volume
  • Cross-reactivity
  • Stability of reagents affects outcome
  • Limited number of analytes in each analysis
  • Proteins
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Safari, F.; Kehelpannala, C.; Safarchi, A.; Batarseh, A.M.; Vafaee, F. Biomarker Reproducibility Challenge: A Review of Non-Nucleotide Biomarker Discovery Protocols from Body Fluids in Breast Cancer Diagnosis. Cancers 2023, 15, 2780. https://doi.org/10.3390/cancers15102780

AMA Style

Safari F, Kehelpannala C, Safarchi A, Batarseh AM, Vafaee F. Biomarker Reproducibility Challenge: A Review of Non-Nucleotide Biomarker Discovery Protocols from Body Fluids in Breast Cancer Diagnosis. Cancers. 2023; 15(10):2780. https://doi.org/10.3390/cancers15102780

Chicago/Turabian Style

Safari, Fatemeh, Cheka Kehelpannala, Azadeh Safarchi, Amani M. Batarseh, and Fatemeh Vafaee. 2023. "Biomarker Reproducibility Challenge: A Review of Non-Nucleotide Biomarker Discovery Protocols from Body Fluids in Breast Cancer Diagnosis" Cancers 15, no. 10: 2780. https://doi.org/10.3390/cancers15102780

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop