Next Article in Journal
Proteomic Characterization of Primary Human Pancreatic Cancer Cell Lines Following Long-Term Exposure to Gemcitabine
Previous Article in Journal
Neurogranin as a Synaptic Biomarker in Mild Traumatic Brain Injury: A Systematic Review of Diagnostic and Pathophysiological Evidence
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

In Search of Ideal Solutions for Cancer Diagnosis: From Conventional Methods to Protein Biomarkers in Liquid Biopsy

by
Anca-Narcisa Neagu
1,*,
Pathea S. Bruno
2,
Claudiu-Laurentiu Josan
1,
Natalie Waterman
2,
Hailey Morrissiey
2,
Victor T. Njoku
2 and
Costel C. Darie
2
1
Laboratory of Animal Histology, Faculty of Biology, “Alexandru Ioan Cuza” University of Iași, Carol I bvd. 20A, 700505 Iasi, Romania
2
Biochemistry & Proteomics Laboratories, Department of Chemistry and Biochemistry, Clarkson University, Potsdam, NY 13699-5810, USA
*
Author to whom correspondence should be addressed.
Proteomes 2025, 13(4), 47; https://doi.org/10.3390/proteomes13040047
Submission received: 3 June 2025 / Revised: 12 September 2025 / Accepted: 17 September 2025 / Published: 23 September 2025

Abstract

Cancer detection has made significant progress, moving from conventional methods to innovative, non-invasive or minimally invasive approaches aimed at improving early diagnosis, precision, and treatment outcomes. This review examines current and emerging diagnostic technologies, including liquid biopsy (LB), molecular biomarkers, and artificial intelligence (AI). LB analyzes biomarkers in bodily fluids, showing promise in detecting tumors at molecular levels, monitoring cancer progression, and predicting treatment responses. The assignment of specific proteoforms, often linked to tumor subtype, stage, and therapy resistance, adds another layer of diagnostic precision, offering valuable insights for personalized oncology. However, the clinical application of LB faces challenges related to sensitivity, specificity, tumor heterogeneity, and a lack of standardized protocols. Relatively high costs, complex result interpretation, and privacy concerns also hinder its widespread adoption in clinical practice. Despite these challenges, advancements in AI, nanotechnology, and multi-omics strategies offer opportunities to enhance cancer diagnostic accuracy. Future developments, including wearable biosensors and lab-on-a-chip technologies, could lead to personalized, real-time cancer detection with improved patient outcomes, potentially redefining cancer care and fostering a more proactive, patient-centered healthcare approach.

1. Introduction

1.1. Challenges of Conventional Cancer Diagnostic

Despite advances in cancer diagnosis, the search for a perfect or ideal method for the detection of malignant tumors remains still elusive. As a result, researchers and clinicians, along with patients actively involved in their care through participatory medicine [1], continue to seek more reliable tools, methods, and solutions. High-quality cancer screening is now focused on the early detection and treatment of precursor lesions, which are essential to reduce morbidity and mortality of cancer [2]. Through earlier cancer detection and with less discomfort, patients can be diagnosed and start treatment sooner, increasing the likelihood of successful outcomes and reducing both the psychological and physical impact of invasive procedures.
Screening and diagnostic tests and/or methods in oncology are diverse and vary depending on the cancer type, stage, and healthcare systems. These methods include conventional imaging techniques (such as X-ray, ultrasound/ultrasonography (US), computed tomography (CT) and magnetic resonance imaging (MRI)), serum tumor biomarker analysis, and the histopathological analysis of tumor tissues. However, these methods still have several limitations, particularly related to sensitivity and specificity [3].
Despite significant advancements in cancer detection, management, and treatment monitoring, one of the major challenges remains the accuracy of screening and diagnostic methods. The effectiveness of these methods depends primarily on their sensitivity and specificity [4,5]. However, screening and diagnostic assessments are prone to errors, and misinterpretation by researchers and clinicians and can lead to serious consequences for both patients and healthcare systems worldwide [6]. In this context, it is crucial to minimize both false-negative and false-positive results, which can lead to overdiagnosis and overtreatment [2].
Sensitivity and specificity are fundamental metrics for assessing the accuracy of a diagnostic test [7]. The term sensitivity (true-positive rate) refers to the probability that a test will correctly identify a positive result when the patient has a certain disease, while specificity measures the probability that the test will correctly identify a negative result when the individual does not have the given disease [8]. Diagnostic accuracy evaluates how effectively a test distinguish between patients with and without the disease [7].
Lipscomb et al. (2022) proposed that an ideal test for screening large populations for multiple types of cancer must achieve clinically optimal sensitivity while minimizing false-positives [9]. For ovarian cancer (OC), Englisz et al. (2024) suggested that an ideal screening test should have a sensitivity greater than 75% and a specificity of at least 99.6% [10]. The reference standard, or gold standard, refers to the best available method or experimental model for reliable identifying a disease, against which the test index results are compared [7].
All methods used in cancer screening and diagnosis have inherent limitations. Conventional approaches are often suboptimal, emphasizing reduced sensitivity, specificity, and accuracy. They can be invasive, limited to certain cancer types, typically detecting one or a few cancer biomarkers. Additionally, these methods are not able to monitor dynamic changes in tumor development and fail to capture the clonal and molecular diversity within tumors. Invasive procedures also carry risks such as infections, bleeding, pain, and other complications related to anesthesia or recovery. Furthermore, they are time-consuming, labor-intense, and resource-consuming, with limited accessibility and high costs. These factors can result in late cancer detection, overdiagnosis, overtreatment, including unnecessary surgery, and delayed treatment decisions. Moreover, interpretation variability and human error can further compromise results.

1.2. Evolution of Cancer Diagnostic Technologies

Although the methods previously discussed remain fundamental in current oncological practice, cancer research continues to explore more precise, non-invasive or minimally invasive, and accessible tools for early and accurate diagnosis. To address these limitations, modern diagnostic methods are increasing being introduced. Cancer diagnostic has evolve from imaging-based techniques, including microscopic detection of malignant lesions, to the sophisticated analysis of tumor biomarkers in vitro and in vivo, using advanced mass spectrometry (MS) techniques, nano-devices, and innovative imaging technologies [11]. Proteoforms are increasingly recognized for their critical role in cancer biology [12]. They provide a more detailed molecular fingerprint of tumors than traditional biomarkers, reflecting the complexity and heterogeneity of cancer at different stages. Detecting proteoforms, particularly through non-invasive liquid biopsy approaches, holds great promise for improving early cancer diagnosis and enabling more precise patient stratification. As technologies for proteoform analysis continue to evolve, they offer valuable opportunities to enhance cancer detection and support the development of personalized treatment strategies. This review examines the evolution of cancer detection methods, from conventional approaches to emerging innovation in liquid biopsy (LB) and molecular biomarkers, with a focus on improving accuracy, minimizing invasiveness, and enabling early detection and personalized treatment.

1.3. The Promise of Liquid Biopsy and Future Directions

LB represents a technique that is transforming the way in which carcinogenesis and cancer development are detected and monitored. LB enables the accurate assignment of tumor-derived biomarkers, offering a less invasive alternative to conventional tissue/surgical biopsies [13]. The term ”liquid biopsy” is defined as a minimally invasive or non-invasive technique, focusing on blood/serum/plasma, urine, saliva, human breast milk, nipple aspirate fluid (NAF), tears, and other body secretions, to analyze tumor-derived molecules circulating in bodily fluids, including protein/peptides, proteoforms, autoantibodies, microRNAs (miRNAs), circulating tumor DNA (ctDNA), metabolites, as well as extracellular vesicles (EVs) and circulating tumor cells (CTCs), which can serve as biomarkers of cancer presence and progression [14]. Furthermore, the sensitive detection of CTCs through advanced platforms like magnetic beads, microfluidic chips or size-sensitive ultrafiltration is gaining attention as an effective biomarker-mediated tool for both cancer diagnosis and prognosis [15]. In particular, early-stage multi-cancer detection, such as for pancreatic, ovarian, and bladder cancers, has shown remarkable potential with a blood test based on EVs proteins, achieving an impressive 71.2% sensitivity and 99.5% specificity [16].
Compared to conventional tissue biopsies, LB has several advantages, including its ability to provide insights into the intratumoral heterogeneity because tumor-derived molecules are shed into bodily fluids from both primary and metastatic sites [17]. Thus, LB offers a comprehensive snapshot of the molecular landscape of cancer, reflecting the genetic and phenotypic diversity of tumors. One of the key advantages of LB is its high sensitivity, reproducibility, and flexibility, making it an attractive option for cancer detection and monitoring [18]. LB allows for repeated testing, enabling real-time monitoring of tumor dynamics, which is particularly valuable for continuous assessment of treatment efficacy, detecting minimal residual disease, and identifying potential relapses in patients.
Proteoforms, different molecular variants of proteins arising from genetic variations, alternative splicing, and posttranslational modifications (PTMs), identified through LBs provide important molecular information that can enhance early cancer detection and treatment [19]. Autoantibodies targeting these altered proteoforms serve as valuable biomarkers, reflecting the body’s immune response to cancer and offering potential for improved diagnosis and novel therapies [20]. A more detailed discussion of proteoforms and their roles is provided in a separate chapter.
LBs contain stable exosomal microRNAs (miRNAs), which represent a promising class of minimally invasive biomarkers for cancer diagnosis and prognosis [21]. These molecules offer valuable insights into disease progression and treatment response across multiple malignancies, including lung cancer, breast cancer (BC), prostate cancer (PCa), oral squamous cell carcinoma (OSCC), colorectal cancer (CRC), and others [21]. However, exosome-based research still faces several challenges, such as the complex, multistep process required from serum collection to miRNA analysis. These include difficulties in isolating tumor-derived exosomes and the lack of standardized methodologies for exosome separation and characterization [21].
In conclusion, LB has emerged as a promising method for detection of several cancer types, particularly in early stages of the disease, where conventional imaging and biopsy methods may fall short. Notably, LB has shown great potential for detecting early-stage BC [22], bladder cancer [18], hepatocellular carcinoma (HCC) [23], pancreatic cancer (PC) [24], and many other malignancies. Thus, LB are a promising tool for cancer diagnosis, prognosis, and patients stratification for personalized therapy [23]. In addition to its diagnostic applications, LB offers unique advantages in identifying therapeutic targets, analyzing drug resistance mechanisms, and monitoring multidrug resistance in advanced cancer patients [25]. Proteome-based plasma tests, for instance, have demonstrated higher accuracy when compared to other technologies and may serve as starting point for developing a new generation of screening tests capable of detecting multiple cancers at an early-stage within general population [26].

2. Limitations of Conventional Diagnostic Paradigms

Despite significant advancements in oncology, conventional diagnostic methods, such as histopathology, imaging, and single-analyte blood tests, continue to face critical limitations, particularly in the early detection of cancer, monitoring minimal residual disease, and capturing tumor heterogeneity. These approaches often rely on invasive biopsies, provide static snapshots of disease, and lack the sensitivity to detect low-abundance biomarkers or early molecular changes. Moreover, they are limited in their ability to guide personalized treatment decisions or adapt to dynamic changes in tumor biology. These shortcomings have underscored an urgent need for more sensitive, specific, and non-invasive diagnostic tools, prompting the development of innovative approaches such as liquid biopsies, multi-omics analyses, AI-assisted diagnostics, and point-of-care (PoC) technologies. These emerging methods aim to overcome the constraints of traditional diagnostics by offering real-time, comprehensive, and minimally invasive insights into tumor biology, thus paving the way for earlier detection, better monitoring, and more precise cancer management. These limitations are discussed in detail in this chapter, highlighting how they have driven the development of more advanced and innovative diagnostic strategies.

2.1. Serum Biomarkers Limitations

Serum and plasma are highly complex mixtures containing thousands of proteins, with some being abundant and others present in much lower concentrations, making detection challenging. Mass spectrometry (MS) play a crucial role in the assignment of cancer biomarkers in human serum due to its ability to detect a wide range of proteins and peptides, including variants [27]. Circulating serum markers, produced by both normal and abnormal cells or tissues, are often considered attractive for clinical use as they can be obtained through minimally invasive methods [23]. While several serum markers have been validated for diagnosis and monitoring cancer progression, none exhibit the necessary specificity, sensitivity, and predictive value required for population screening [27].
For instance, prostate-specific antigen (PSA), alpha-fetoprotein (AFP), cancer antigens such as CA-125, CA-15.3, CA-19-9, and carcinoembryonic antigen (CEA) are still commonly used for detecting prostate, liver, ovarian, breast, pancreas, or colon cancers, respectively, despite their limitation in accuracy and early-stage detection [27].
PSA is sensitive but poorly specific for PCa detection in symptomatic patients, often leading to overdiagnosis and treatment of latent cancer without clinical evidence. This is due to its inability to distinguish between indolent and aggressive forms of PCa [28,29]. Serum PSA is an example of a widely used biomarker for screening, diagnosis, and prognosis of PCa. However, it is considered an organ-specific biomarker, even if a study of nipple aspirate fluid (NAF) found that women with no risk factors of BC had high levels of PSA, while women with precancerous or invasive lesions had reduced PSA levels [30]. PSA is non-cancer specific and its levels can also be elevated in non-cancerous conditions, such as urinary tract infection, prostatitis and benign prostatic hyperplasia (BPH) [31,32].
Furthermore, serum PSA can be influenced by a variety of internal and external factors. To address these limitations, several fluid-based molecular biomarkers have been developed for PCa detection [33]. One such biomarker is the Prostate Health Index (PHI), which combines total PSA, free PSA, and [-2] proPSA (p2PSA) isoform into a score derived from a simple, inexpensive blood test [34]. PHI has been shown to improve detection of high-grade PCa compared to PSA-based assays alone [35]. Another example is the 4Kscore blood test, which integrates four kallikrein protein biomarkers (total PSA, free PSA, intact PSA, and human kallikrein-related peptidase 2) into an algorithm to enhance the prediction of aggressive form of PCa. This allows men with low 4Kscore results to safely delay biopsy [36]. The prostate cancer antigen 3 (PCA3), which is highly expressed in prostatic tumors, can be measured through a urine test. The PCA3 test offers acceptable sensitivity and specificity, making it a valuable non-invasive method for PCa diagnosis [37].
Alpha-fetoprotein (AFP) is a glycoprotein that is often elevated in the sera of patients with liver cancer [38]. However, elevated serum AFP have also been reported in patients with other cancers, including BC, esophageal, cervical, pancreatic, endometrial, gastric, lung, and rectal cancers [38]. Additionally, AFP can be elevated in non-cancerous conditions, such as cirrhosis, nephrotic syndrome, and gastritis [38]. Similarly, serum CA-125 is an extensively used biomarker for OC diagnosis. However, CA-125 levels may also be elevated in other malignant and non-cancerous conditions, such as endometrial, breast, pancreatic, and gastrointestinal cancers, as well as liver cirrhosis, endometriosis, and large uterine fibroids/leiomyomas [10,39,40]. Furthermore, CA-125 levels were found to be significantly higher in patients with polycystic ovarian syndrome (PCOS) compared to control patients, with a value of 11.45 U/L showing high sensitivity (96.7%) and specificity (87.1%) for PCOS [41]. This suggests that CA-125 could be used as an additional diagnostic biomarker for this disease [41]. Additionally, Kim et al. (2015) reported that higher plasma volumes in obese patients could dilute serum tumor biomarkers such as PSA for PCa, and CA-125 and CA-19-9, which can affect the efficiency of tumor screening for ovarian and pancreatic cancer [42]. Despite these challenges, serum CA125 has shown the highest sensitivity (93.30%), while human epididymis protein 4 (HE4) has demonstrated the highest specificity (97.87%) in diagnosis of epithelial ovarian cancer (EOC) [43]. Additionally, CA-125 and HE4 are clinically approved biomarkers for the diagnosis and prognosis of endometrial cancer (EC) [44]. In these cases, the combined detection of serum soluble epidermal growth factor receptor (sEGFR), CA-125, and HE4 has been shown to increase the specificity and efficiency in EOC diagnosis [43].
Carcinoembryonic antigen (CEA) was originally identified as a colon cancer antigen, and elevated serum CEA levels have led to its clinical use as a diagnostic biomarker for CRC [45]. However, CEA levels are also elevated in cancer such as pancreatic and rectal cancers, as well as in non-cancerous conditions like lung fibrosis, uremia, chronic obstructive pulmonary disease (COPD), and Alzheimer’s disease [45]. Furthermore, CEA levels are known to increase with aging [45].
For early cancer detection, risk assessment, and therapy-related monitoring, improving serological biomarkers across different cancer types is essential [46]. LB offers significant advantages over conventional serum biomarkers due to its ability to capture a broader spectrum of molecular information, including mutations and PTMs of proteins that may be overlooked by conventional serum markers. As a result, LB can provide a more comprehensive understanding of tumor mechanisms and support the development of personalized treatment strategies. Therefore, advances in innovative proteomics technologies are urgently needed to facilitate biomarker discovery in tumor-associated LBs [47].
High-plex proteomics technologies, ranging from MS and antibody/antigen arrays to innovative approaches like aptamer-mediated LB [48], proximity extension assay (PEA), and reverse phase protein arrays (RPPA), are capable of simultaneously measuring hundreds of proteins in LB samples and can detect cancer at early stage [47]. These technologies exploit highly accurate and unbiased affinity-based biomolecule capture of from LB, primarily relying on nucleic acid aptamers, synthetic DNA or RNA oligonucleotides with unique tertiary structures that selectively target proteins and other molecules. Aptamers leverage the specificity and binding affinity of ligands toward surface receptors on circulating targets [48,49]. For detecting novel cancer biomarkers, affinity-based methods enable large-scale proteome analysis in complex bodily fluids, overcoming the limitations of conventional, and even advanced, MS-based proteomics [46]. Roy et al. (2021) and other authors demonstrated that nucleic acid aptamers exhibit high specificity and affinity, as well as modifiability and flexibility, making them easily adaptable for evolution and functionalization. These characteristics make aptamers a reliable alternative to antibodies for diagnostics, imaging, and targeting applications, positioning them as ideal recognition elements for LB [48,50]. Monoclonal antibody production is more expensive than the process of fabricating and improving aptamers, which can be successfully used for immunohistochemistry and LB [51]. Consequently, aptamer-based recognition of BC cells opens a new era for BC diagnosis [51]. High-multiplex aptamer-based serum proteomics can accurately discriminate between patients with OSCC and healthy controls, achieving 100% accuracy. This approach demonstrates that panels of OSCC-associated proteins revert to levels seen in healthy controls after tumor resection, identifying candidate cancer-specific serum biomarkers [46]. Moreover, aptamer-based microfluidic platforms or chips for detecting CTCs and EVs hold great potential as powerful tools in clinical applications [52]. Aptamers have emerged as cost-effective detection tools for targeting HE4, which is overexpressed in OC, making them a promising strategy for OC diagnostics or the development of urine biosensor-based tests for OC [53]. Additionally, the development of cancer-specific smart aptamers enables the targeted delivery of therapeutic molecules directly to tumor cells, minimizing damage to healthy cells and tissues [54,55].
In conclusion, despite significant progress in the discovery and validation of serum biomarkers for cancer, traditional approaches based on canonical proteins remain limited by issues of specificity, sensitivity, and clinical utility. As shown with markers such as PSA, AFP, CA-125, and CEA, many of these proteins are elevated not only in malignancies but also in a variety of benign or unrelated conditions, leading to overdiagnosis, false positives, and poor predictive value, particularly in early-stage disease. These limitations stem from a gene-centric view of the proteome, where proteins are treated as direct correlates of gene expression, ignoring the functional diversity generated by alternative splicing, PTMs, and proteolytic processing. In reality, proteomes are composed of proteoforms, distinct molecular species of proteins that represent the actual biological effectors within cells and tissues. Therefore, effective biomarker discovery must move beyond protein-level assignement toward proteoform-level resolution, where these functional differences can be captured and quantified [56].
Conventional MS-based approaches, especially bottom-up methods, often fall short in capturing this complexity. Bottom-up (BUP) MS enables high-throughput protein assignment but lacks the resolution needed to distinguish proteoforms, isoforms, and complex PTMs due to peptide-level limitations [57]. However, integrative top-down proteomics (iTDP) has emerged as the only analytical strategy currently capable of routinely detecting and quantifying the broad diversity of proteoforms in native biological samples [58]. While iTDP offers the necessary depth and resolution, it comes with trade-offs, such as higher complexity, cost, and reduced throughput compared to faster, lower-resolution methods.
Biomarker discovery stands at a crossroads: prioritizing speed and scalability often comes at the cost of accuracy and biological relevance, while rigorous, high-resolution analyses better capture the complexity of the proteome. Although slower, proteoform-level data provide deeper insights and more selective biomarkers. High-plex technologies like aptamer arrays and PEA expand coverage but must maintain high analytical standards [59]. Embracing proteome complexity is essential for advancing precision oncology in the post-proteogenomic era [56].

2.2. Histopathology Limitations

Histology, including histopathology, immunohistochemistry (IHC), electron microscopy (EM), and immunocytochemistry (ICC), plays a crucial role in clinical diagnosis in oncology. However, there are cases where histology alone cannot provide a definitive diagnosis, requiring multidisciplinary approaches that involve expert teams for accurate results [60,61]. To enhance tumor detection and management, molecular biomarkers and genetic analysis are essential in complementing histopathological findings with molecular diagnostics [61].
Histological analysis typically requires a biopsy to obtain a tumor tissue sample, a procedure that can be invasive and associated with complications such as infections, bleeding, or pain. This is particularly problematic for patients undergoing invasive biopsy procedures, such as core needle biopsies or surgical biopsies, which necessitate longer recovery times. Additionally, biopsy may not be feasible or accessible in certain cases, such as with deep-seated tumors, a high risk of bleeding, or multiple tumor sites [62]. Conventional histopathological tissue diagnosis methods are also time-consuming and labor-intensive, often taking several days, which can delay both diagnosis and treatment decisions [63].
This delay can be particularly problematic in urgent cases where rapid decision-making is crucial. Additionally, a biopsy samples only a small portion of the tumor tissue, which may not be representative to the entire tumor, especially given that tumors are often characterized by high genetic and molecular intraheterogeneity. In such cases, a biopsy may fail to capture the full molecular diversity present within tumor, potentially compromising the accuracy of the histopathological diagnosis. Moreover, early lesions or small tumors may be missed, affecting the detection of early-stage cancer or micrometastases.
Histological analysis typically provides a snapshot of the tissue at the time of the biopsy but does not offer a prospective outlook on tumor progression [64]. To monitor tumor changes over time or detect recurrence, multiple biopsies may be required, which can be invasive and, in some cases, impractical. Additionally, the use of 2D tissue sections can limit the ability to analyze the tumor in its three-dimensional context. However, recent advances in microscopic and computational technologies now enable successful 3D reconstruction of tissues and tumors, improving the scope of analysis [64]. In certain types of cancer, histology alone may not provide sufficient diagnostic or prognostic information.
Although histopathology is a crucial diagnostic tool, widely regarded as the ”gold standard” for cancer diagnosis, biopsy results can still be variable, often subjective, and semi-quantitative [65,66]. As a result, a second opinion from an additional pathologist is often sought [66]. Tseng et al. (2023) found that seeking a second opinion can lead to changes in the diagnosis, with shifts from benign to malignant diagnoses in 10–19% of cases [66]. Additionally, partial or minor disagreement regarding the grade or subtype of the tumor occurred in 20–34% of cases, while significant changes in prognosis or the treatment occurred in 17–39% of cases [66]. The same authors also highlighted that interpreting of histological margins of tumors, especially when tissue processing is inadequate, presents a challenge for pathologists. These interpretations can be time-consuming and, at times, subjective [66].
It is estimated that 85% to 95% of breast lesions biopsied show benign histology, and the costs associated with these procedures can be considerable [67]. For BC detection, fine needle aspiration cytological biopsy (FNAC/FNAB) and core needle biopsy (CNB) are commonly used prior to surgical biopsy and are generally considered relatively inexpensive diagnostic procedures [68]. Other minimally invasive sampling methods have become more preferred, including ultrasound guided FNAB (US-FNAB) and image guided CNB (IG-CNB) [67]. A systematic review and meta-analysis by Wang et al. (2017) evaluating suspicious breast lesions concluded that CNB has better sensitivity (95%) compared to FNAC (74%), although the specificity of CNB is similar to FNAC (98% vs. 96%) [69]. Silva et al. (2023) reported that the sensitivity, specificity, and accuracy of FNAC (97%, 94%, and 95%, respectively) were comparable to CNB (97%, 96%, 96%, respectively) as CNB, with FNAC showing lower complication rates [68]. A comparative high-throughput proteomic analysis of matched CNB, FNAC, and surgical resection tissue samples from BC patients revealed that about 85% of proteins were identified in all sample types, indicating that FNAC is suitable for proteomic analysis and holds promise as a source for assignment new protein biomarkers for BC [70]. Several proteins, including cyclin D1 (CCND1), cyclin-dependent kinase (CDK6), HER2, and insulin-like growth factor 1 receptor (IGF1R), were found in higher quantities in FNAC compared to tissue samples, while tubulin beta 2A (TUBB2A) was detected exclusively in FNAC [70].
Immunohistochemistry (IHC) is an essential method for detecting specific diagnostic proteins within tissue samples, but it remains limited to detecting one biomarker per tissue section or, in dual-color IHC assays, two biomarkers simultaneously [71,72,73]. While multiplexed immunofluorescence (IF) can visualize multiple proteins at once, it is constrained by spectral overlap. These challenges can be addressed using Fluorescence Lifetime Multiplexing (FLEX), which allows for the differentiation of 11 or more biomarkers in 3D tissues [71]. Despite being a qualitative and subjective approach, IHC continues to be a powerful tool for detecting the in situ distribution of biomarkers in FFPE tissue sections. However, IHC also has several limitations, including a lack of standardized guidelines for staining, which leads to variability in results across different laboratories due to the use of different antibodies or protocols [74].
For instance, the accurate and reliable assessment of human epidermal growth factor receptor type 2 (HER2) status, along with other hormone receptors (HRs) such as estrogen receptor (ER) and progesterone receptor (PR), is crucial for stratifying cancer subtypes and determining appropriate treatments. However, the inter-assay accordance for detecting HER2-low tumors is not high. Even with the same staining assay, various factors can influence the result of HER2 IHC-based detection [75,76].
Consequently, it is critical to accurately identify HER2-low cancer in HER2-negative patients [76]. Studies have shown that factors such as the type of fixative used, as well as the time to and duration of fixation of tumor tissues can impact HER2 staining intensities and FISH scores [75,76]. Additionally, changes in antigen retrieval and antibody incubation procedures can affect staining intensity of HER2 expression [76]. A significant discordance in HR positivity rates has been reported between IHC analysis (84%) and enzyme immunoassay (EIA) (45%) in invasive lobular carcinoma, concluding that IHC is more sensitive, specific, and cost-effective than EIA [77]. Accurate evaluation of HER2 status is essential for patient stratification and precision therapy, and IHC remains one of the most commonly used pathological method for HER2 status [65]. In this context, Thanasan et al. (2023) recommended using IHC alone for patients with BC undergoing anti-HER2 treatment, even without in situ hybridization (ISH), due to the strong concordance between HER2 IHC scores and HER2 amplification, with a sensitivity of 87.96%, specificity of 93.75%, and an accuracy of 89.74% for BC diagnosis [78]. Furthermore, a meta-analysis by Shamshirian et al. (2020) revealed serum HER2 levels have low sensitivity (53.05%) but reasonable specificity (79.27%) and an accuracy of 72.06% for BC diagnosis. This suggests that assessment of serum HER2 levels can serve as a valuable verification test [79].
Molecular characterization is crucial for a better understanding of tumor characteristics [80]. Mass spectrometry imaging (MSI) is as an excellent method for the accurate detection and mapping of tissue biomarkers in oncological applications [80], and it is a powerful tool for investigating tumor aggressiveness based on cancer biomarkers [81]. Additionally, MSI allows for the analysis of histopathological features without the need of specific antibodies [80]. Using MSI-based techniques, Gonçalves et al. (2023) demonstrated that vimentin (VIM) overexpression is associated with triple-negative breast cancer (TNBC), a finding validated by IHC, suggesting that VIM could be a promising target for BC diagnosis and treatment [80].
These histology-related limitations underscore the need for complementary technologies, such as LB or advanced imaging techniques, to enhance cancer screening, detection, and management. The main advantages of LB vs. histopathology are listed in Table 1.

2.3. Imaging Methods Limitations

The majority of breast tumors are initially detected by patients through breast self-examination (BSE). Although recommendations suggest that women should continue performing BSE, even if its efficacy remains controversial, as research has not clearly linked BSE with significant benefits in early detection [82]. Studies have shown that BSE has low estimated sensitivity (20% to 30%), and its effectiveness tends to decrease among older women [83]. BSE remains one of the first steps women take in detecting abnormalities of the breast.
While clinical breast examination (CBE) performed by an experienced health examiner is the next option, it also has limitations in sensitivity. An alternative is the Intelligent Breast Exam (iBreastExam or iBE), which electronically palpates the breast to identify possible abnormalities. This device has been shown to have significantly better sensitivity than CBE (63% vs. 31%) [84]. Recent studies, such as by Bhimani et al. (2023), demonstrated that iBE, a portable, wireless, radiation-free, cost-effective, easy-to-use, and hand-held device, could be a valuable tool for BC detection. Sensitivity with iBE ranges from 34.3% to 86%, and specificity ranges from 59% to 94% [85]. This device has moderate to higher diagnostic ability for malignant lesions, with a sensitivity for identifying breast tumors ranging between 57% and 93%, allowing for the identification of breast tumors ranging between 0.5 cm to 9 cm [85].
Imaging plays a critical role in BC diagnosis, with mammography being the gold standard. However, ultrasonography (US) is increasingly emerging as a routine alternative or complementary imaging method [86]. Sood et al. (2019) demonstrated that US can be an effective primary detection tool for BC, especially in low-resources settings or where cost is a consideration. The overall sensitivity and specificity of US were reports to be 80.1% and 88.4%, respectively [87]. US is particularly beneficial for BC diagnosis in women with dense breast tissue, where other imaging methods may be less effective [87].
When it comes to imaging methods for BC screening, X-rays are used in both film mammography and digital mammography [88]. However, film mammography has limited sensitivity in detecting BC, particularly in women with dense breast. In these cases, digital mammography offers significant improved accuracy [89]. For instance, in a study involving Japanese women aged 40–49, the sensitivity of modern film mammography alone was found to be 44.1% for women with dense breasts and 34.8% for women with non-dense breasts [90]. Digital mammography has a sensitivity of 97%, specificity of 64.5%, and diagnostic accuracy of 89.3%, making it an accurate tool for detecting BC. However, mammography is not always feasible or available, particularly in certain regions or settings [87,88]. In 2015, in the Journal of the Royal Society of Medicine, Gøtzsche argued that mammography screening could be more harmful than beneficial. The author suggested that it does not increase the women’s lifespan, instead contributing to overdiagnosis and leading to unnecessary mastectomies [91]. According to Gøtzsche, if mammography-screening were a pharmaceutical drug, it would likely have been withdrawn from the market due to its negative effects [91].
Computed tomography (CT) is a widely used non-invasive imaging modality that plays a crucial role in the diagnosis of various diseases, including cancer [92]. Despite the fact that CT scans expose patients to ionizing radiation, which can potentially lead to carcinogenic mutations and DNA damage, they remain an invaluable tool in cancer diagnosis worldwide [92,93]. A meta-analysis involving 111.6 million adult participants from Europe, America, and Asia found a dramatically increase in cancer risk associated with CT scans, with the risk being positively correlated with the radiation dose and the CT sites [93]. The long-term effects of radiation exposure can manifest over several decades [92]. However, some studies indicate that exposure to multiple CT scans and other diagnostic radiologic imaging (10–20 scans) with a cumulative dose of 100-200 mSv does not significantly increase cancer risk [94].
A recent study evaluating 144 cases of CT scans correlated with histopathological findings showed a sensitivity of 98.52% and specificity of 87.50% for CT scans. The precision and accuracy of CT scans were reported as 99.25% and 97.91%, respectively [95]. In the context of lung cancer screening, low-dose CT demonstrated a sensitivity of 88.9% and specificity of 92.6%, while chest X-ray had a sensitivity of 78.3% and specificity of 97.0% [96]. These findings indicate that low-dose CT screening for lung cancer has higher sensitivity but slightly lower specificity compared to conventional chest X-rays [96]. In another study analyzing 142 pediatric whole-body MRI results, the sensitivity for detecting malignancies was 93.8%, with a specificity of 93.4%. The positive predicted values was 65.2%, while the negative predicted value was 99.1% [97]. In BC diagnosis, Aziz et al. (2022) examined the concordance between radiological and histopathological findings. They reported a sensitivity of 94.48% and specificity of 43.14% for Breast-Imaging and Reporting Data System (BI-RADS) in relation to mammography and breast US, highlighting the significant correlation between radiological and histopathological results in invasive BC [98].
F-18 fluorodeoxyglucose (FDG) PET/CT has been shown to have superior diagnostic performance compared to conventional imaging methods for detecting distant metastases in BC. Specifically, FDG PET/CT exhibits a higher sensitivity (97.4%) and specificity (91.2%) compared to conventional imaging, which has sensitivity and specificity of 85.9% and 67.3%, respectively [99]. In the context of detection of head and neck squamous cell carcinoma (HNSCC), the latest-generation FDG-PET/CT demonstrates improved sensitivity in detecting cervical lymph node (LN) metastasis, while specificity remains similar or slightly lower than older PET/CT scanners [100].
Prostate-specific membrane antigen (PSMA) is a type II transmembrane glycoprotein typically expressed in the cytoplasm of normal prostate cells. It is overexpressed on almost all PCa cell membranes, and this overexpression is correlated with increased DNA repair mutations and the development of hormone-resistant PCa [101]. PSMA PET/CT, a next-generation imaging modality, has significantly improved the clinical diagnosis of PCa across various clinical settings. It offers greater sensitivity and specificity compared to conventional imaging methods such as CT, MRI, and bone scintigraphy [102,103].
However, one of the challenges with PSMA PET/CT is limited number of specialists trained to interpret these scans, and there is variability in data interpretation among different readers [104]. To address these challenges, AI-based techniques can be employed to assist in the interpretation of PCa imaging. These AI methods can aid in detecting primary tumors, interpretation, including detection of primary tumor, identifying local recurrences, assessing metastatic lesions, classifying lesions, and even predicting or prognosticating disease outcomes [104].
LB has emerged as a crucial tool in the management of PCa, using prostatic secretions, urine, and blood as sources for biomarker discovery, validation and clinical implementation [33,105]. Numerous urinary biomarkers have been proposed for PCa detection, enhancing diagnostic accuracy and patient monitoring [106]. A study by Höti et al. (2023) demonstrated that urinary PSA, when combined with serum PSA, exhibited a higher predictive ability in differentiating PCa from other conditions [107]. The authors found that patients with aggressive PCa had lower levels of urinary PSA compared to those with non-aggressive cancer. In contrast, serum PSA levels were significantly higher in patients with aggressive cancer [107]. Additionally, the study revealed that aggressive PCa was negatively correlated with PSA expression in PCa tissues as analyzed by IHC [107].
In the current era of proteomics, mass spectrometry (MS)-based technologies, such as matrix-assisted laser desorption/ionization MS (MALDI-MS), surface-enhanced laser desorption/ionization MS (SELDI-MS), and multiplexed isobaric tagging technology for relative quantitation (iTRAQ), enable for detection and quantitative analysis of the PCa proteome. These technologies are shifting the focus from a single biomarker (typically PSA) to panels of protein biomarkers, which necessitate the discovery of novel biomarkers for improved diagnosis and patient stratification [108]. It is well known that a single biomarker, while providing its own sensitivity and specificity, is insufficient for accurately diagnosing and stratifying PCa [29]. Consequently, the use of multiple biomarkers can enhance diagnostic accuracy compared to individual biomarkers [29].
In a study by Franzi et al. (2023), capillary electrophoresis coupled with mass spectrometry (CE-MS) profiling was used to develop a multi-dimensional biomarker model that included 181 urinary peptide biomarkers, which demonstrated strong accuracy in detecting PCa [106]. Many of these peptides were derived from collagen proteins, indicating the involvement of extracellular matrix in tumor invasion and progression. Other biomarkers included proteolytic products of proteins such as S100A9, uromodulin, albumin, fibrinogen alpha, mucin-2, beta-2-syntrophin, alpha-1-acid glycoprotein 1, protein phosphatase 1 regulatory subunit 3A, while markers like gelsolin, insulin-like growth factor II, and prostaglandin-h2-isomerase were found to be decreased in the urine of PCa patients compared with non-PCa group [106].
Additionally, using liquid chromatography coupled with mass spectrometry (LC-MS) to profile endogenous peptides in urine samples from patients with PCa, benign prostate hyperplasia (BPH), and healthy individuals, De Souza Dutra et al. (2023) identified five urinary peptides derived from uromodulin that are less abundant in the urine of PCa patients and might be considered as putative PCa biomarkers, able to differentiate between PCa and BPH [29]. These urinary peptides were shown to have high sensitivity (81.82%) and specificity (88%), outerperforming PSA in distinguishing between PCa and BPH [29]. The main advantages of LB vs. computed tomography (CT) are listed in Table 2.

3. What’s Innovative in Cancer Detection and Management?

Innovation in cancer detection and management lies in the integration of advanced technologies and methodologies that enhance early diagnosis, improve accuracy, and minimize invasiveness. These alternatives to conventional methods enable the non-invasive detection of cancer, offering a real-time snapshot of tumor genome, transcriptome, proteome, and metabolome. They also enable early identification of cancer even before clinical symptoms appear. Advanced imaging technologies offer greater precision in detection of malignancy at the molecular level, improving early diagnosis and correct staging. Artificial intelligence (AI) and machine learning (ML) are increasingly integrated into diagnostic platforms, analyzing vast amounts of medical data, which expand rapidly due to advances in digital health tools and analytics. In this context, the use of nanotechnologies has emerged as a complementary innovation, offering new possibilities for improving diagnosis, treatment, and monitoring in oncology. Together, these innovative approaches (Figure 1) will be reviewed in the following subchapters, highlighting their significant potential to transform the landscape of cancer detection, and lead to better treatment decisions and personalized care for each patient.

3.1. Established Clinical Standards as a Foundation for Innovation

While established clinical standards, such as multimodal imaging (e.g., CT, MRI, PET), targeted molecular assays, and histopathology remain the cornerstone of cancer diagnosis and management due to their validated clinical utility, they are increasingly being augmented by innovative approaches. These novel methods aim not to replace, but to complement and enhance current diagnostic workflows, enabling earlier detection, better stratification, and more precise therapeutic interventions.
  • Multimodal imaging
To enhance diagnostic accuracy across various cancer types, the combination of different imaging techniques has been revolutionary, providing a more precise and comprehensive view of tumors. The latest-generation 18F-fluoro-2-deoxy-glucose positron emission tomography/computed tomography (18F-FDG PET/CT) offers improved sensitivity and image quality compared to older PET/CT scanners [100]. Furthermore, due to its higher sensitivity, the 18F-FDG PET/CT scan plays a synergistic role with MRI in diagnosing sub-centimeter malignant lesions [109]. The combination of PET and MRI in PET/MRI hybrid technology offers distinct advantages over PET/CT, including the absence of ionizing radiation, enhanced soft-tissue contrast, a wide range of acquisition sequences, and potential superiority in specific body areas and specific situation, such as tumor recurrence detection [110]. For instance, whole-body 18F-FDG PET/CT imaging has limitations in detecting brain metastasis form non-central nervous system tumors due to high background activity, making MRI scanning a valuable complementary tool [111]. Emerging combinations with artificial intelligence (AI) or molecular probes are pushing multimodal imaging toward next-generation diagnostics, offering enhanced sensitivity and precision.
  • Genetic and genomic testing
Genetic biomarkers are playing an increasingly important role in early cancer detection, prognosis assessment, and treatment planning or management, supporting the development of targeted therapies aimed to improve patient outcomes. Thus, genetic testing, which analyzes gene expression profiles and mutations, has become an important tool in improving the accuracy of cancer diagnosis and guiding personalized treatment strategies [112]. The decision between single/limited gene testing and multigene panels depends on clinical context [113]. For example, in early-stage BC, multigene assays (MGAs) are utilized to support decisions regarding adjuvant therapy and to help estimate patient prognosis, while for individuals with metastatic BC, testing for specific genetic alterations can identify therapies with potential efficacy [112]. For instance, multigene prognostic genomic assays have become essential in managing early-stage BC [114]. Notable examples include the Oncotype DX Recurrence Score® (RS), also known as the 21-gene recurrence score assay, widely used in Europe and America [112,115], MammaPrint® (MMP), also known as the 70-gene risk of distant recurrence signature [112,116], ProsignaTM, a PAM50-based subtype classifier and risk model [117], EndoPredict®, a 12-gene molecular score (MS) [118], and Breast Cancer Index (BCI), a gene-expression based assay that evaluates tumor proliferation pathways and estrogen signaling in BC [119]. The NanoString nCounter® DX Analysis System offers a digital profile of up to 800 genes, providing more precise and accurate measurements of mRNA expression levels in formalin-fixed paraffin-embedded (FFPE) specimens compared to PCR-based methods [117]. Tissue-based gene expression tests also increasingly used to estimate PCa aggressiveness [120]. Recently, McHugh et al. (2025) published a study about the assessment of a Polygenic Risk Score in screening for PCa [121].
Thus, the advent of next-generation sequencing (NGS) technology allows multigene testing for hereditary cancer risk [113]. Current clinical cancer genome sequencing typically relies on targeted exome panels, which focus on protein-coding regions representing only a small fraction of the genome [122]. As a result, somatic mutations in non-coding regions—such as regulatory elements, non-coding RNAs, and mitochondrial DNA—remain largely unexplored [122]. With technological progress, emerging applications, such as fragmentomics, a new field studying fragments of circulating DNA (cirDNA) for cancer screening, are gaining attention [123]. Circulating tumor DNA (ctDNA) has emerged as a promising tool for cancer detection, prognosis, and treatment monitoring through liquid biopsy [124]. This approach captures tumor-derived DNA from both primary and metastatic sites and allows for the detection of genetic and epigenetic alterations [124]. Additionally, structural variants and viral integrations in cancer genomes are not yet comprehensively studied [122]. To solve these inconvenient, the appropriate use of genomic testing, such as whole-genome sequencing (WGS), whole-exome sequencing, or multigene tumor profiling, has become increasingly important in modern oncology [125]. Unlike traditional genetic testing, which primarily focuses on identifying germline mutations linked to cancer risk, genomic testing provides a comprehensive analysis of somatic alterations within the tumor itself [125]. This enables more precise diagnosis, prognosis, and selection of targeted therapies, making it a key tool in the era of personalized medicine.
WGS is known as a powerful NGS tool with large applications in cancer research field [126]. WGS enables a wide range of analyses, including detection of non-coding mutations (e.g., in promoters and regulatory regions), large structural variants (deletions, insertions, inversions, duplications, translocations), viral genome integration, mitochondrial alterations, and immune-related features like HLA typing [122]. It also supports the diagnosis and treatment of hereditary cancers through germline variant analysis, pharmacogenomics, and mutational signature profiling [122]. Whole genome and transcriptome sequencing (WGTS) provides superior diagnostic and therapeutic insights compared to gene panel testing by detecting a broader range of relevant genetic alterations [127]. It enables prediction of tissue-of-origin in most previously undiagnosed cases and informs treatment decisions for a higher proportion of patients. Additionally, WGTS applied to cell-free DNA shows promise as a feasible method for guiding clinical management [127].
The disadvantages of genetic and genomic testing include the ethnic disparities that may impact MGA testing outcomes [112]. Moreover, there are limitations to genomic testing, primarily related to their high costs, which can range from 2000 US$ to 4000 US$ [116,125]. Although ctDNA shows potential as a biomarker in various cancers, challenges remain due to its low concentration, short half-life, and the lack of standardized cut-off values [124]. A major challenge is expanding the use of NGS to all cancer patients to support informed treatment decisions [128]. Key concerns include tissue sample quality, preservation, and health technology assessment [128]. WGS also has limitations, such as shallow read depth—risking false negatives in highly heterogeneous tumors—the need for frozen tissue, large data volumes, high analysis and storage costs, and difficulties in interpreting novel variants [122]. These challenges are being addressed by long-read sequencing technologies, which improve structural variant detection, and deep learning methods like convolutional neural networks, which enhance variant interpretation [122].
Genetic test misinterpretation is common and can affect clinical decisions [129]. Challenges often involve unclear reports, limited counseling, and variants of unknown significance. Improved communication and further research are needed to reduce errors. In conclusion, genomic testing can improve cancer care by guiding risk assessment, prognosis, and therapy, yet its use remains limited due to difficulties in result interpretation and lack of clear guidance for non-geneticists [130].
  • Liquid biopsy sources for detection of protein biomarkers
LB are increasingly used alongside traditional biopsies, offering the advantage of detecting disease progression months before it becomes visible through imaging techniques or clinical evaluation [21]. Detection of proteins and their diverse proteoforms, including autoantibodies, in LBs offers valuable insights into tumor biology, serving as sensitive and specific biomarkers for early cancer diagnosis, disease monitoring, and personalized treatment strategies.
Over the past decades, significant efforts have been dedicated to exploring, discovering, and validating novel, sensitive, specific, and accurate protein biomarkers for cancer diagnosis, prognosis, and targeted therapy [131]. Investigating tumor protein biomarkers offers several advantages over conventional cancer detection techniques, such as serological markers, biopsy, and imaging. The combinatorial benefits of tumor protein biomarkers include their non-invasive or minimally invasive nature when assessed through body fluids, like LB, enabling early-stage cancer detection. Additionally, they provide high capacity for monitoring treatment responses, guiding personalized and targeted therapies, and supporting precision oncology. Protein biomarkers are also valuable for detecting minimal residual disease, reducing radiation exposure, enabling risk stratification, and improving quantification and prediction. Their broad applicability extends to rare cancers and tumors located in difficult-to-assess organs, offering a cost-effective and less discomforting alternative for cancer patients.
Proteoforms detected through LB provide critical, detailed molecular insights that enhance early cancer diagnosis, enable more precise disease monitoring, and support personalized treatment strategies. Autoantibodies targeting tumor-associated proteoforms represent a particularly valuable class of biomarkers, reflecting the body’s humoral immune response to altered self-proteins, such as mutated, aberrantly expressed, or post-translationally modified autoantigens [20]. This immune recognition often occurs early in tumorigenesis, making autoantibodies useful not only for diagnosis and monitoring, but also for identifying druggable targets and informing novel immunotherapeutic approaches [20]. A study conducted by Montero-Calle et al. (2023), highlights the potential of using different proteoforms of the p53 and p63 proteins as early blood-based markers for CRC [19]. By analyzing how the immune system reacts to these specific protein variants in blood samples, researchers found that certain proteoforms were more commonly recognized in patients with CRC or precancerous lesions than in healthy individuals. The results showed high accuracy in distinguishing these groups, and testing with user-friendly biosensor devices confirmed their potential for clinical use. These findings suggest that p53 and p63 proteoforms could help improve early detection of CRC through a simple blood test [19]. Moreover, autoantibodies targeting p53 are frequently associated with CRC, owing to the high mutation rate of the TP53 gene in this malignancy [20]. While these autoantibodies exhibit high specificity in advanced CRC (89–100%), their diagnostic sensitivity remains limited (8.8–46.3%). Notably, emerging evidence suggests that p53 autoantibodies can also be detected in early-stage CRC and premalignant lesions, albeit with variable sensitivity (10–45.2%). Furthermore, autoantibodies against other p53 family proteoforms, such as p73, ΔNp73α, and ΔNp73β, have shown enhanced diagnostic performance [20]. These proteoforms may become immunogenic due to their dysregulation or mutation in CRC. Recent studies report that autoantibodies against these variants achieve sensitivities above 49%, specificities exceeding 86%, and strong discriminatory power for early detection (AUC > 65%), including in premalignant conditions [20].
LBs offer a promising platform for the non-invasive detection of PTMs of proteins, enabling dynamic monitoring of disease-related signaling alterations and treatment responses in real time. Phosphorylation, a key protein PTM, often becomes dysregulated in cancer, leading to abnormal signaling and therapy resistance. To address this, Ahmed et al. (2023) developed a multiplexed phosphoprotein analyzer chip (MPAC) capable of rapid and sensitive detection of phosphorylated proteins in lung cancer [132]. Using lung cancer cell lines and patient-derived EVs, the chip profiled major pathways (MAPK and PI3K/AKT/mTOR) and detected treatment responses, including to kinase inhibitors and immunotherapy [132]. A phosphorylation heat map from plasma samples distinguished cancer from noncancer individuals and revealed proteins associated with therapy response. This tool shows strong potential for personalized cancer treatment through dynamic phosphoproteomic monitoring.
The proteomics-based workflow for the detection of protein biomarkers in LB (Figure 2) relies widely on high-resolution liquid chromatography-tandem mass spectrometry (LC-MS/MS), a powerful platform for both discovery and targeted proteomics [133,134].
This process typically begins with the collection of biofluidsamples such as plasma, serum, urine, tear biofilm, breast milk, NAF, or saliva, followed by protein extraction and enzymatic digestion—commonly using trypsin—to generate peptide mixtures. These peptides are then separated via liquid chromatography (LC) and analyzed by tandem mass spectrometry (MS/MS), enabling precise identification and quantification of proteins. LC-MS/MS offers high sensitivity and specificity, allowing for the comprehensive profiling of the proteome, including low-abundance proteins and critical PTMs of proteins that may serve as cancer biomarkers [135]. Quantification can be performed using either label-free approaches or chemical labeling techniques such as isobaric tags for relative and absolute quantitation (iTRAQ, TMT), which enhance throughput and enable comparative analyses across multiple samples. This workflow is particularly well-suited for non-invasive liquid biopsies, as it facilitates the identification of disease-associated protein signatures that differentiate between healthy and pathological states. Moreover, LC-MS/MS supports both untargeted (discovery-phase) and targeted (validation-phase) proteomics, making it instrumental in biomarker discovery, early cancer detection, real-time monitoring of disease progression, and assessment of therapeutic response. By delivering deep proteome coverage with robust analytical performance, LC-MS/MS-based proteomics is paving the way for the development of clinically actionable biomarker panels that can significantly enhance the precision and personalization of cancer diagnostics and monitoring.
Due to the high complexity and variability of the human proteome, there is a growing demand for innovative technologies that enable accurate and efficient biomarker discovery [47]. Recent advances in MS instruments, such as the Q Extractive HF, Orbitrap Exploris 480, timsTOF Pro 2/HT, and ZenoTOF 7600—have dramatically improved scan speed, resolution, and dynamic range, enhancing detection and quantification of low-abundance proteins [136].
A major advancement is this field is the integration of ion mobility technologies, such as high-field asymmetric waveform ion mobility spectrometry (FAIMS) and trapped ion mobility spectrometry (TIMS). These methods provide additional gas-phase separation, reduce background noise and increase proteome coverage, especially in complex clinical samples like FFPE tissues and low plasma volumes [137]. This improvement supports more comprehensive profiling of the proteome and proteoforms, including dysregulated species with biomarker potential. Using timsTOF-based four-dimensional label-free proteomics, Lu et al. (2025) successfully enhanced the detection of low-abundance proteins in patient-derived EVs [138]. They found that EVs from metastatic nasopharyngeal carcinoma (NPC) patients were significantly enriched compared to those from locoregional NPC and healthy individuals, and were functionally involved in processes like cell motility, metabolism, and angiogenesis [138].
Furthermore, the parallel reaction monitoring-parallel accumulation-serial fragmentation (prm-PASEF) method on the Bruker timsTOF Pro mass spectrometer enables highly sensitive, multiplexed, and absolute quantification of plasma proteins [139]. By integrating ion mobility, this technique improves depth and speed of analysis, quantifies proteins across a wide dynamic range, and shows excellent concordance with traditional multiple reaction monitoring (MRM), making it particularly suitable for clinical biomarker validation [139]. These technological advances in MS and ion mobility significantly enhance biomarker discovery by enabling deeper, faster, and more sensitive proteome analysis, especially from complex samples. They hold strong potential for advancing early cancer detection and precision medicine through LB applications.
The main sources for LB include a variety of bodily fluid such as blood/plasma/serum, urine, saliva or oral fluid, human breast milk, nipple aspirate fluid, pleural fluid, and aqueous humor. The following sections examine each of these liquid biopsy sources, outlining their specific advantages, biomarker content, and relevance to cancer diagnostics.
  • Blood/Serum/Plasma
A recently published editorial written by Bradley and Watson (2025) showed that blood tests was the most frequently performed category in patients who were later diagnosed with cancer, 44% of patients receiving such a test [140]. PCa is the most common urological cancer in men worldwide, substantially contributing to morbidity and mortality [106,108,141]. For PCa diagnosis, the serum PSA, an important biomarker that correlates with the risk of PCa, and digital rectal examination (DRE) are the most commonly used methods, followed by biopsy of the prostate gland, considered the gold standard diagnostic technique for PCa detection [107,142]. In primary care for PCa detection in England, the PSA level obtained after asymptomatic PSA testing is useful in diagnosing one in five patients with PCa [89]. However, PSA is unable to distinguish between indolent and aggressive PCa [29]. Merriel et al. (2022) showed that PSA is highly sensitive but poorly specific for PCa detection on symptomatic patients [28]. The DRE sensitivity performed by primary care clinicians during a study conducted by Naji et al. (2018) was 51%, with 59% specificity [141]. Serum PSA is an appropriate example of serum biomarker largely used as a screening test in the diagnosis and prognostication of PCa that is organ –but not cancer-specific, its level being also elevated or extremely elevated in non-cancerous prostatic conditions, including urinary tract infection, prostatitis and benign prostatic hyperplasia (BPH) [31,32]. Streicher et al. (2019) revised the sensitivities reported for serum PSA in different studies, reporting a sensitivity of 32% for PCa detection, using a threshold value of 3.1 ng/mL, and 21% sensitivity for 4 ng/mL, leading to overdiagnosis and overtreatment of latent cancer without clinical evidence, correlating to a relatively high rate of unnecessary biopsies [142].
Furthermore, it is important to note that the level of serum PSA can be influenced by various internal and external factors. Thus, there are studies indicating a correlation between age and serum PSA levels in different populations [143,144]. Serum PSA level of obese men is lower than similarly aged lean men, due to hemodilution and hormonal changes based on an higher estradiol/testosterone ratio [145]. Moreover, Koc et al. (2013) showed that cigarette smoking affects hormone levels, so the PSA level could be higher in smokers compared with non-smokers [146]. Press et al. (2020) reported that cigarette smoking and tobacco use were associated with an increase in PSA among older African American men in Chicago, whereas marijuana use was inversely associated with PSA levels [147]. Ethnic group-based differences in PSA levels in men without PCa have been reported [148]. Thus, Black men have higher PSA levels than White or Hispanic men, resulting in higher PCa incidence and mortality in Black men than White men, while Asian men tend to have lower PCa incidence and mortality than White men, but the effects of ethnicity on PSA level are not clearly demonstrated [148,149].
Blood LB could be a valid alternative to tissue biopsy in PCa for diagnostic use, molecular profiling, as well as for discovery of predictive and prognostic biomarkers [17]. There is a pressing need for improved biomarkers to aid clinicians in diagnosis, prognostication, assessment of therapy response, and characterization of mechanisms of resistance [150].
  • Urine
Urine is a complex biological fluid that is proven to give valuable insights into the physiological and pathological processes of the human body. As a filtrate of blood plasma, it contains a wide variety of biomolecules originating from the whole organism. Urine contains a wide range of proteins, including those secreted by the cells of kidneys or the urinary tract (70%), as well as those present in the systemic circulation at the time of filtration (30%) [151]. The use of proteins from urine samples for cancer detection and identification has gained significant attention in the past years. This strategy takes advantage of proteomic analysis using methods such as mass spectrometry (MS) to identify reliable biomarkers for various cancers, including breast (BC), bladder and PCa. Urine has some advantages and disadvantages. It can be collected in significant quantities, in a simple and non-invasive manner, without causing much discomfort to patients. This property of urine sampling makes it a preferred choice for the early-detection and long-term monitoring of cancer [152], as well as for individuals requiring frequent assessments. It has been shown multiple times that the proteome present in urine reflects local and systemic physiological changes in the body. Thus, its analysis can be used to assess a wide range of biomarkers related to cancer or other diseases, being especially promising for the investigation of hard-to-biopsy tissues [153]. On top of this, up-to-date proteomic techniques such as MS and proximity-extension analysis (PEA) have enabled the cost-effective and high-throughput analysis of the urinary human proteome. These methods have improved both the qualitative and quantitative identification of novel cancer biomarkers, making them well-suited for large scale clinical studies [154].
However, there are some drawbacks worth taking into consideration when using the analysis of urinary proteins as a tool for disease diagnosis and monitoring. It is well known that urine samples are characterized by very high intra- and inter-individual variability due to factors such as lifestyle, dietary patterns and renal function. This inconsistencies can slow the identification of new biomarkers and reduce the reliability of diagnostic tests [152,155].
Moreover, it is very likely that more diseases, if present in the same individual, can impact the same urinary biomarkers in different ways, thus hindering the result of these analyses. The low protein concentration of urine represents another disadvantage, making it difficult to identify and accurately quantify low-abundance biomarkers [156]. Techniques such as data-independent mass-spectrometry (DIA-MS) has shown promising results on overcoming this limitation [154]. Additionally, urine is known to contain high levels of urea, salts, and other interfering molecules that can hinder urine proteome analysis [152,157]. This limitation requires additional preparation steps that can make a clinical study more time-consuming and less cost-effecting. In addition, lack of standardized protocols for urine sample collection, processing and analysis is likely contributing to inconsistencies in study results, thus hindering the translation of experimental biomarkers into clinical settings [158].
A recent study by Kong et al. (2024) set out to identify novel bladder cancer biomarkers, revealing 40 significantly altered serum proteins and 17 urine proteins compared to healthy controls. Among them, ESM-1, AREG, RET, WFDC2, FGFBP1 and PVLR4 were successfully validated using singe-cell sequencing and IHC. Using LASSO regression, a 14 protein diagnostic model (11 from serum and 3 from urine) was created. This model achieved high-diagnostic accuracy, outperforming FISH in the detection of the early-stage and low-grade bladder cancer [18]. Another study investigating urinary proteins as cancer biomarkers successfully identified specific heat shock proteins (HSPs) with distinct expression patterns across various cancer types using machine learning (ML). HSP90AB1, TRAP1, FKBP4, HSPA9, and HSPB5 were identified to present unique signatures in urine that correlate with the presence of bladder, lung, and other cancers. Notably, low CCT2/HSP90AB1 levels were associated with non-cancer groups, while low HSP90AB1/TRAP1 and HSPA6/TRAP1 were associated with an increased chance of cancer presence. Moreover, this study also underscores the fact that isoforms of HSPs could help differentiate benign lung disease from lung cancer, reinforcing the diagnostic potential of HSP profiles in cancer detection and identification [159]. Several biofluids contain prostate-derived EVs and reports suggest that EVs from the bloodstream can be found in urine [160]. Urinary LB is emerging as a promising method for PCa detection [161]. Exosomes, as a novel type of liquid biopsy, have the potential to become diagnostic and prognostic biomarkers [162].
These up-to-date findings highlight the potential of urinary proteomics in cancer diagnosis. By harnessing advanced proteomic techniques and machine learning, urine-based liquid biopsies could offer a non-invasive, cost-effective, and sensitive approach to cancer detection and monitoring.
  • Saliva or oral fluid
Saliva or oral fluid, is a clear and odorless biofluid secreted by the salivary glands, playing a crucial role in maintaining homeostasis within the oral cavity [163]. It is composed predominantly of water approximately (99%), alongside a diverse array of proteins (including enzymes, mucins, and immunoglobulins), other organic molecules (such as glucose, nucleic acids, lipids, and metabolic waste products) and electrolytes [163,164,165]. The term total human saliva refers to the final composition of this secretion, which also incorporates crevicular fluid- an inflammatory exudate derived from periodontal tissues, along with cellular debris, upper airway secretions, and microorganisms residing in the environment [166].
Salivary composition is subject to high intra- and inter-individual variability. Among the factors contributing to this variability, salivary flow rate appears to be the most influential [163]. Similarly to urine, saliva offers a major advantage in clinical applications due its easy, safe, and non-invasive collection process, making it especially suitable for repeated sampling required in cancer monitoring [167]. In recent years, saliva has emerged as a promising medium for detecting breast cancer (BC) [168] or oral and head and neck cancers [169] an others. For instance, a systematic review revealed that saliva, as a non-invasive biomarker, has the potential to accurately differentiate between BC patients from healthy controls with a pooled specificity and sensitivity of 72.7% and 71.7%, respectively [168]. Some of salivary constituents have been identified as potential biomarkers for these malignancies. A study by Krapfenbauer et al. (2014) identified a list of proteins detected by two-dimensional polyacrylamide gel electrophoresis (2DE-PAGE) in human saliva, including keratins, keratin subunits, enzymes and enzyme inhibitors, cytokines, immunoglobulins as well as amylase and other salivary specific glycoproteins, some of them significantly overexpressed in OSCC [170]. The molecular profile of saliva also carries valuable physiological and pathological information, particularly relevant to the head and neck region, potentially offering insights into cancer development and progression [171].
In addition to its diagnostic potential, saliva-based testing is cost-effective, making it a compelling option for large-scale screening initiatives. Furthermore, it poses minimal risk of cross-contamination and is safer for healthcare-workers [172]. However, despite these advantages, saliva-based liquid biopsies still face notable challenges. Chief among them is the lack of standardized protocols for sample collection, processing, and analysis, which undermines the reliability and reproducibility of biomarker identification [173,174]. Even if a real-time, non-invasive diagnostic tool for oral cancer were available, implementing a widespread screening program might still be inefficient due to the relatively low incidence of these cancers [172,174]. Moreover, salivary samples can be easily contaminated depending on various factors, such as the timing of food intake, oral bleeding, or enzymatic activity from either the host or oral microbiota. These variables can alter biomarker stability and interfere with diagnostic accuracy [172].
A recent systematic review by Bastías et al. (2024) evaluated 62 studies to identify promising salivary biomarkers for the early detection of oral carcinoma. All included studies reported high specificity and sensitivity of the proposed biomarkers [175]. This analysis covered over 60 biomolecules found in saliva samples from more than 5000 subjects, including patients with OSCC, oral potentially malignant disorders (OPMDs), and healthy controls. The authors highlighted TNF-α, IL-1β, IL-6, IL-8, LDH, and matrix metalloproteinase 9 (MMP-9) as particularly promising biomarkers for both OSCC and OPMDs. However, they showed that it is unlikely for a single biomarker to serve as a definitive diagnostic tool. Supporting this perspective, a study conducted by Ghallab and Shaker (2016) demonstrated that both chemerin and MMP-9 are significantly elevated in the serum and saliva of patients with OSCC [176]. Notably, the study found that salivary chemerin and MMP-9. As well as serum chemerin, were highly effective in distinguish OSCC patients from those with oral premalignant lesions (OPMLs). These biomarkers exhibited 100% sensitivity and 100% specificity, outperforming serum MMP-9 in diagnostic accuracy [176].
The authors advocate for the development of biomarker panels, which can more accurately reflect the complex physiological and pathological states associated with malignancy [175]. Future research should aim to validate these findings by developing and testing robust biomarker panels. Such efforts should include comparative analyses of multiple body fluids and assess the sensitivity and specificity of biomarker combinations for improved early cancer detection and monitoring.
  • Human breast milk
Breast milk proteome analysis emerged as a promising method for assessing BC risk, as it provides access to molecular profiling for a high volume of breast tissue [13,177,178]. Recent research has focused on identifying possible biomarkers in breast milk that could help in the early detection, diagnosis and monitoring of BC. Mass spectrometry-based proteomics is one of the most accurate methods for the analysis of breast milk samples due to its ability for identification and quantification of proteins, but also of their isoforms, variants, and modified proteins [177]. Thus, one-dimensional (1D) SDS-PAGE (separation based on molecular weight as the only dimension) or two-dimensional (2D) SDS-PAGE (separation based on molecular weight and isoelectric point as the two dimensions), as well as 1D or 2D polyacrylamide gel electrophoresis (1D PAGE and 2D PAGE) coupled with nano-liquid chromatography-tandem MS (nLC-MS/MS) lead to accurately identification of dysregulated proteins that play a role in cancer progression and might be considered as putative BC biomarkers [179]. A series of studies revealed differential expression of certain proteins in the breast milk of BC-positive women vs. healthy controls, such as lactadherin, also known as milk fat globule-epidermal growth factor 8 (MFG-E8) or secreted protein containing EGF-like repeats and discoidin/F5/8 complement domains (SED1), lactoferrin (LF), gelsolin (GSN), alpha-amylase, vitronectin (VTN/VN), casein family proteins, albumin, etc. [177,179,180,181]. Lactadherin (MFG-E8) plays important roles in cell adhesion, angiogenesis promotion, and tissue regeneration, emerging as a biomarker of BC, and associated with poor prognosis and decreased survival not only in BC, but also in ovarian, colorectal, melanoma and other cancer types [182]. LF is an ubiquitous multifunctional iron-binding glycoprotein of the transferrin family, present in various biological fluids, such as milk, pancreatic secretion and bile, saliva, as well as in neutrophils [183,184]. LF, detected as downregulated in breast milk of BC women [185] could play a role in tumor promotion, especially in cancers of the female reproductive system and BC, mainly contributing to development of TNBC phenotype [184]. Keskin et al. (2022) reported that calprotectin (CP) and LF levels increased significantly during radiotherapy in the mouth rinse samples of patients with head- and neck-cancer, but their levels decreased significantly after the treatment, suggesting the radiation-therapy-induction of inflammatory environment in the oral cavity [186]. GSN, a calcium-regulated actin-binding protein, has the ability to be a prognosis, diagnostic, and immune biomarker in pan-cancer, mediating cytoskeletal remodeling and regulating the epithelial-to-mesenchymal (EMT) transition [187]. Evidence suggests that GSN had a good diagnostic value in serum of BC patients [187]. Alpha-amylase, upregulated in breast milk of BC women [185], was also found as significantly overexpressed in saliva of patients with oral cancer [188], and emphasized inhibitory effects on cancer cell proliferation and glucose uptake in human neuroblastoma cell lines [189]. VTN, a multifunctional adhesive glycoprotein involved in cell growth, cell adhesion, angiogenesis, ECM degradation and metastasis, was found as upregulated in breast milk of BC patients [185], but it was assessed as overexpressed in patients with metastatic undifferentiating neuroblastoma [190]. Although these putative biomarkers show promise in detecting BC, they lack the specificity to be exclusive to a single cancer type. They have been dysregulated in the development and progression of multiple malignancies, suggesting that they could be part of a pan-cancer biomarker profile. Evidence suggests that the proteomic profile of breast milk could complement other screening methods, especially for young women with dense breast tissue.
  • Nipple aspirate fluid (NAF)
NAF is a natural secretion produced by the luminal epithelial cells that lines breast ducts, which can be non-invasively acquired via nipple by aspiration using a suction device, resulting in a liquid biopsy from both breasts. Thus, NAF may recapitulate the characteristics of the microenvironment of breast tumor [191]. Proteomic patterns of NAF can be obtained by use of surface enhanced laser desorption and ionization time of flight (SELDI-ToF) MS that is rapid and reproducible, identifying the differentially expressed proteins (DEPs) from BC patients compared to healthy controls, including those with abnormal mammogram but biopsy normal [192]. Thus, NAF is also a promising source of biomarkers for BC detection, emerging as an additional tool next to imaging or as a replacement tool for when application of imaging methods is not possible [193]. Many tumor biomarkers have been investigated in NAF, such as CEA and PSA [193]. As a future perspective, NAF biomarker profiling could be used for routine breast health monitoring by home-based liquid biopsy collection kits [30].
  • Pleural fluid (PF)
PF can be considered as a successful source of liquid biopsy and is an useful opportunity especially in cases where tissue biopsy is not available [194]. Thus, malignant pleural effusion (MPE) arises during progression of many tumors, such as lung, breast and ovary cancer [194,195]. MPF are richer in tumor-derived products that can be clinically exploited than plasma [194].
  • Tear fluid
Tears are a clear extracellular fluid secreted by the lacrimal glands, containing a diverse array of mRNA, lipids, and relatively high concentrations of proteins/peptides, including enzymes and cytokines, that could be considered as putative cancer biomarkers [196]. Thus, tears reflect systemic and local physiological changes, offering clinically relevant information from various bodily tissues [197]. Most proteins identified in plasma were found in tear samples, suggesting that tear fluid may serve as a surrogate for blood in certain diagnostic contexts [198]. A study by Daily et al. (2022) demonstrated the utility of tears as a non-invasive source for the early detection of breast cancer (BC), utilizing LC-MS/MS for protein profiling, followed by ELISA for biomarker validation [197]. These findings support the potential role of tear-based testing as a complementary tool to conventional screening methods, such as mammography [199]. Kaufmann et al. (2022) showed that protein profile in tears of patients with CRC could have a real potential as a non-invasive screening test, compared with invasive colonoscopy or other screening options with low sensitivity as the fecal immunochemical test [200].
In addition to proteomic analysis, human tears have also been explored as a source for studying PTMs, which may offer further insights into disease processes at the molecular level [201]. The non-invasive nature of tear collection, combined with minimal sample processing requirements and short processing times, makes tears a particularly attractive biofluid for diagnostic applications [198]. However, before tear-based testing can be adopted in clinical practice on a large scale, standardization of procedures related to sample collection, storage, and protein extraction, similar to those established for blood, is essential [198].
  • Aqueous humor (AH)
In addition to commonly used biofluids, AH is increasingly recognized as a valuable source for proteomic analysis, particularly in ocular cancers like uveal melanoma (UM) and retinoblastoma (Rb), where it can provide insights into tumor biology and support minimally invasive diagnostic strategies. AH analysis in eyes affected by UM revealed significantly elevated levels of inflammatory, angiogenic, and immune-related biomarkers, such as IL-6, IL-8, CCL5/RANTES, EGF, basic fibroblast growth factor (bFGF), macrophage migration inhibitory factor (MIF), and MCP, compared to controls [202]. A borderline increase in VEGF was also observed. Notably, IL-8 levels correlated with tumor thickness, and IL-6 with serous retinal detachment, suggesting a link between biomarker concentration and tumor characteristics. These findings highlight the potential of AH proteomics to characterize the UM tumor microenvironment and contribute to improved understanding and monitoring of the disease [202]. Rb, the most common intraocular cancer in children, traditionally cannot be biopsied due to risk of tumor spread. However, recent advances in treatment now permit safe extraction of AH, offering a novel opportunity to use AH as a liquid biopsy [203]. AH biomarkers may now play a clinical role in diagnosis, prognosis, and disease monitoring during therapy. This shift marks a potential breakthrough in non-invasive Rb management [203].

3.2. Near-Clinical or Translational Tools

In recent years, a growing number of innovative cancer detection tools have moved beyond the proof-of-concept stage and are now being rigorously evaluated in clinical research settings. While not yet fully established in routine clinical practice, they are increasingly integrated into clinical trials and translational studies, reflecting their potential to complement or even transform existing diagnostic workflows. Their continued development and validation mark a critical step toward more precise, non-invasive, and real-time cancer care.
  • Pan-cancer analysis and multi-omics approaches
Pan-cancer analysis represents a transformative strategy in oncology, enabling the identification of molecular patterns shared across different tumor types. Pan-cancer analysis, in conjunction with multi-omics strategies, facilitates cancer diagnosis by identifying patterns common to multiple cancer types, thereby leading to personalized and effective therapies [204]. For example, Sharma et al. (2024) showed that comprehensive multi-omics analysis of BC integrates proteomics, transcriptomics, and metabolomics data obtained from tumor tissues, revealing distinct long-term prognostic subtypes [205].
The Clinical Proteomic Tumor Analysis Consortium (CPTAC) has generated extensive proteomic datasets by analyzing tumor samples, previously studied by The Cancer Genome Atlas (TCGA), using advanced MS [206]. These datasets support integrative proteo-genomic research by linking protein and genomic data, particularly in colon, breast, and ovarian cancers. To ensure consistency across analyses, CPTAC developed the Common Data Analysis Platform (CDAP), which standardizes data processing from raw spectra to peptide and gene-level outputs [206]. The platform performs peak extraction, database searching, and false-discovery filtering, while also offering phosphosite localization via PhosphoRS program. Outputs are provided in accessible formats, enabling cross-sample and cross-cancer comparisons [206].
Using reverse-phase protein arrays (RPPA) as a valuable tool in functional proteomics for analyzing signaling pathways involved in cancer, researchers have profiled approximately 8000 samples from 32 cancer types within The Cancer Genome Atlas (TCGA), leading to the development of The Cancer Proteome Atlas (TCPA), an open-access resource [207]. To enhance its usability, a new “TCGA Pan-Cancer Analysis” module was introduced, allowing integrated, protein-focused analyses across cancers. This includes evaluating relationships between protein expression and genetic alterations, epigenetic changes, mRNA, and miRNAs, as well as building protein co-expression networks and identifying clinically relevant biomarkers [207].
Additionally, protein PTMs are critical regulators of cellular signaling in both healthy and cancerous cells. Using high-throughput MS, researchers analyzed PTM profiles in over 1100 patients across 11 cancer types through CPTAC [208]. The study uncovered shared patterns of protein acetylation and phosphorylation linked to key cancer pathways, such as DNA repair, immune response, and histone regulation. It also revealed interactions between PTMs that influence kinase activity. These findings deepen the understanding of cancer biology and may offer new therapeutic targets [208].
Such integrative approaches enable the development of precision medicine strategies, where the therapy can be tailored to the molecular profile of individual patients, rather than relying solely on conventional clinical parameters. Furthermore, pan-cancer analysis holds the potential to uncover shared bio-pathological pathways and biomarkers across different cancer types, which could lead to the development of broad-spectrum therapies.
  • Artificial intelligence (AI) and machine learning (ML)
Standard AI methods for histopathology analyses have revolutionized cancer diagnosis by optimizing specialized models for each diagnostic task, aiding in both cancer detection and prognosis [209]. AI and ML technologies are increasingly employed to analyze medical imaging, histopathology slides, and panels of tumor biomarkers, while also incorporating patient history. This integration allows AI and ML to identify diagnostic and prognostic patterns that enhance the accuracy of conventional diagnostic methods. AI and ML are particularly effective in reducing human error, enabling faster, more consistent, and holistic interpretation of large and complex datasets. As large-scale datasets continue to be generated across different studies, these innovative technologies play a crucial role in managing the vast amounts of data, something conventional statistical methods often struggle with. This capability is especially important in the discovery of complex biomolecular relationship, where ML and AI can uncover patterns that might not be apparent through manual analysis [210].
Moreover, the integration of AI and ML into multi-omics approaches holds significant potential for improving early cancer detection and monitoring. By combining multiple biomarkers from different bodily fluids, AI-powered algorithms can enhance the sensitivity and specificity of cancer screening, enabling the cancer detection earlier, more treatable stages [211].
The versatility of AI and ML algorithms (MLAs) is exemplified by their broad applications in oncology. Various algorithms, such as neural networks, logistic regression, random forest, and support vector machine, have demonstrated promising results in cancer diagnosis, medical imaging analysis, personalized treatment, and even drug discovery [212,213]. These ML-based models have the ability to process and learn from vast, multi-dimensional data, facilitating the assignment of novel biomarkers, optimization of treatment strategies, and prediction of patient outcomes. As AI and ML continue to evolve, their integration into clinical workflows could pave the way for more precise, efficient, and patient-specific care.
By analyzing large datasets, ML and AI algorithms can identify molecular signatures that distinguish cancer patients from healthy controls, as well as detect biomarkers that are crucial for accurate cancer diagnosis [214,215,216]. For example, Qureshi et al. (2024) explored the use of AI and ML in profiling pancreatic ductal adenocarcinoma (PDAC) and discovering biomarkers, focusing on the connection between distinct proteomic and metabolomics urine biomarkers of PDAC and their diagnostic value [215]. Their study found that the XGBoost classifier outperformed conventional statistical techniques, achieving 89% accuracy and 91% sensitivity in diagnosing PDAC [215]. ML has been employed to identify novel diagnostic biomarkers for non-small cell lung cancer (NSCLC) and to identify new therapeutic targets for this type of cancer [216].
Taking it a step further, MLAs trained on panels of biomarkers specific to certain cancers can detect subtle abnormalities in biomolecule expression patterns, even at early stages, making early diagnosis more feasible [217]. Moreover, ML-based models have the potential to predict an individual’s likelihood of developing a specific cancer, underscoring the importance of ML as a powerful tool in cancer diagnostics [217].

3.3. Advanced Proteomic Methods with Growing Translational Promise

  • Protein isoforms as precision biomarkers
Distinct protein isoforms, often arising from alternative splicing of RNA and specific PTMs of proteins, can serve as highly specific biomarkers for cancer detection and chemotherapy resistance. They enable the differentiation between tumor subtypes or stages and support more personalized therapeutic strategies [12]. Ligand binding assays remain the common methods for detecting proteoforms in clinical laboratories; however, these tests can miss unexpected modifications, making MS essential for accurate analysis of proteoforms in biological samples [12].
For example, p95 HER2, a truncated isoform of HER2 found in about 30% of HER2-positive BCs, is associated with more aggressive disease and resistance to the monoclonal antibody trastuzumab [218,219]. Lacking the extracellular domain, p95HER2 escapes antibody binding while maintaining oncogenic activity, making it a potential biomarker for poor prognosis and treatment response [218]. CD44, a cell surface glycoprotein, is overexpressed in most epithelial tumors and is a key marker for cancer stem cells. Specific CD44 variant isoforms play crucial roles in tumor growth, metastasis, and chemotherapy resistance by activating oncogenic pathways [220]. Targeting these variants shows promise for enhancing both cancer diagnosis and therapy. In pancreatic ductal carcinoma (PDAC), aberrant isoforms of CA125/MUC16 were associated with aggressive tumor subtypes and are implicated in disease progression and metastasis [221]. These findings highlight the clinical relevance of MUC16 isoforms as both biomarkers of disease aggressiveness and therapeutic targets in PDAC.
In conclusion, the characterization of distinct protein isoforms has become increasingly important for improving cancer diagnosis, predicting treatment response, and guiding personalized therapies. Advances in detection methods, particularly mass spectrometry, have enhanced the ability to identify clinically relevant isoforms, which are associated with tumor aggressiveness and therapeutic resistance. Continued research and clinical validation of these isoform-based biomarkers hold promise for advancing precision oncology and minimizing unnecessary interventions.
  • Mass spectrometry imaging (MSI)
MSI is an advanced, label-free technique that enables the spatial mapping and quantification of molecules directly within tumor tissue samples, providing detailed molecular information without the need for labels or tags [222]. Moreover, MSI has emerged as a powerful technique for the spatially resolved detection of proteoforms, including clinically relevant isoforms, directly within tissue microenvironment (TME) [223]. A recent multiplexed workflow called ”AutoPiMS”, a semi-automated top-down tandem mass spectrometry (MS2) approach, demonstrates the feasibility of high-throughput proteoform assignment at single-ion sensitivity [223]. Applied to human OC tissue, AutoPiMS enabled identification of 73 proteoforms up to 54 kDa at a rate of less than one minute per proteoform [223]. MSI integration revealed over 300 differential proteoforms between tumor and stroma, highlighting intratumoral heterogeneity. Fourteen, including methylated cysteine-rich protein 1 (CRIP1) isoforms, were spatially validated at 20µm resolution [223]. These findings highlight how spatially resolved proteoform analysis can reveal cancer-specific molecular signatures and support the development of tissue-contextualized biomarkers, offering new opportunities for precision oncology and isoform-driven diagnostics.
MSI offers several advantages over conventional diagnostic methods, such as histopathology or immunohistochemistry (IHC). It provides a much comprehensive and nuanced view of tumor characteristics, allowing clinicians to assess not only the presence of specific tumor antigens but also their precise localization within the tissue architecture. This ability is crucial in understanding the molecular complexity of cancer, which influence tumor behavior, metastasis, and treatment responses.
  • Single-cell proteomic analysis
Mass spectrometry and microscopy-based single-cell proteomic profiling are designed to assess intratumoral heterogeneity. These innovative approaches are expected to provide novel prognostic biomarkers and facilitate the development of personalized treatments, thus advancing both cancer detection and therapy [224,225].

3.4. Innovative, Emerging Clinical Tools

  • MasSpec Pen technology
To overcome histology-related limitations, the innovative MasSpec Pen technology leverages a small volume of water droplets in contact with the tissue surface for molecular extraction, followed by mass spectrometry analysis for molecular profiling of diagnostic proteins, lipids, and metabolites. This technology provide a rapid, accurate, and non-destructive diagnostic approach for human cancer tissues or tumor margin assessment during surgery, or ex vivo, in various cancer types, including pancreatic cancer (PC) [226], OC [227], BC [228] as well as lung and thyroid cancers [63]. Although MasSpec Pen is not strictly a liquid biopsy, it shares several similarities with LB techniques due to its ability to analyze a variety of biomarkers in tissue samples quickly and non-invasively. The MasSpec Pen is an automated, biocompatible, and handheld MS device that generates mass spectra leading to the identification of potential cancer biomarkers. It allows for cancer prediction with high sensitivity (96.4%), specificity (96.2%), and overall accuracy (96.3%), in addition to predicting different subtypes of lung cancer and differentiating benign from malignant thyroid tumors [63].
In case of PC, the MasSpec Pen demonstrated a concordance with histopathological results of 91.5%, with a sensitivity of 95.5% and specificity of 89.7% for differentiating normal pancreas from cancer [226]. For distinguishing bile duct from pancreatic cancer, MasSpec Pen achieved an overall accuracy of 95%, with a sensitivity of 92% and specificity of 100% [226]. Sans et al. (20219) demonstrated that the linear ion trap mass spectrometer could actualy discriminate between high-grade serus carcinoma of the ovary and normal ovarian tissue, achieving 100% sensitivity and 100% specificity [226]. In BC, Garza et al. (2024) reported a 95.5% agreement with postoperative histopathology results [228].
  • Nanotechnology in cancer molecular diagnosis
Nanotechnology plays a crucial role in cancer molecular diagnosis by enabling the development of nanomaterials that can selectively recognize tumor-associated protein biomarkers, along with other molecules, and facilitate their visualization [11]. For example, liganded nanoclusters, as nonlinear optical contrast agents, can be used for molecular diagnostics in cancer, enhancing sensitivity and specificity of detection [11]. Nano-machines, as nanoscale mechanical devices, are able to detect, capture, and identify cancer biomarkers, or function as targeted drug delivery systems (DDSs). These devices have wide-ranging applications in cancer diagnosis and imaging, drug delivery, theranostics, and nano-surgery [229].

3.5. Future Point-of-Care (PoC) and Multiplexing Tools

The development of PoC diagnostics has emerged from advances in nanotechnology and nanomaterials. These have enabled for non-invasive, faster, easier, and more cost-effective testing, providing in-site measurements with high sensitivity and selectivity, thanks to progress in biosensor-based technologies driven by innovative nanomaterials, like metal nanoparticles, carbon nanotubes, and graphene, able to enhance sensor performance for cancer biomarker detection at ultra-low concentrations [230,231,232]. Today, nanotechnologies facilitate the real-time detection of tumor biomarkers, offering great potential for early cancer diagnosis, personalized treatment strategies, and effective cancer monitoring. As the design and functionality of nanomaterials continue to evolve, the accuracy, sensitivity, specificity, and versatility of various biosensors, such as optical nanomaterials, are expected to enhance the quality of life for cancer patients [233].
Recent advances in precision medicine rely on innovative technologies that are affordable, simple, and versatile enough to detect multiple molecular biomarkers in real time from various biofluids [234]. Electrochemical bioplatforms detect cancer-related biomarkers by converting biological interactions (e.g., with DNA, RNA, proteins, or metabolites) into electrical signals [232]. They offer rapid results, minimal sample preparation, and high sensitivity, enabling early-stage cancer detection.
Electrochemical biosensor technologies became promising tools for integrating multi-omics data to support advanced diagnostics, therapy, and personalized oncology. Electrochemical biosensors have shown significant promise in the multiplex detection of BC biomarkers, surpassing traditional clinical methods like ELISA, FISH, and PCR in sensitivity [235]. These multiplexed electrochemical sensing platforms enable simultaneous detection of proteins (e.g., HER2, MUC1, and CA 15-3), miRNAs (e.g., miR-155, miR-21, and miR-16), exosomes, and cytokines (e.g., RANKL and TNF) at ultra-low concentrations, often outperforming conventional diagnostic thresholds [235]. For instance, miRNA biomarkers can be detected at femtomolar levels, while proteins such as EGFR and VEGF are measurable at picogram levels—well below ELISA detection limits. Despite their analytical superiority, clinical adoption remains limited, raising key questions about sensor integration, miniaturization, preferred bioreceptors, and regulatory hurdles. This highlights the need for continued research to translate these innovations into PoC tools for early and multiplexed BC diagnostics [235]. Recently, Dosnon et al. (2025) proposed a wearable in-pad diagnostic named MenstruAI, a microfluidic diagnostic platform integrated into menstrual hygiene pads for non-invasive, pain-free monitoring of menstruation blood-based biomarkers [236]. It enables naked-eye or smartphone-assisted detection of several biomarkers such as C-reactive protein (CRP), carcinoembryonic antigen (CEA), and cancer antigen 125 (CA-125), showing strong correlation with venous blood results [236]. This affordable and accessible technology offers a promising tool for early disease detection and improved women’s health monitoring, potentially democratizing healthcare and increasing diagnostic equity.
The main challenges for clinical implementation of biosensors include: improving selectivity in complex biological environments, miniaturizing devices for potential implantable applications, integrating data analytics and machine learning for accurate interpretation, addressing biomarker variability and validation, navigating regulatory pathways and ensuring safety, and overcoming toxicity concerns and commercial scalability [231]. Addressing these challenges requires strong interdisciplinary collaboration and innovation across research, clinical, and regulatory sectors.

4. Biomarkers: Challenges and Limits

While LB represents a transformative approach in oncology, its clinical utility is often limited by the non-specific nature of certain biomarkers. Molecules such as CP, although elevated in various malignancies, are also significantly expressed in a range of non-cancerous inflammatory and infectious conditions. This overlap can lead to false-positive interpretations and complicate the differentiation between cancer-related and benign processes. The reliance on non-specific biomarkers, particularly when used alone, underscores the pressing need for more refined panels that combine sensitivity with specificity, as well as the importance of interpreting LB results within a broader clinical and diagnostic context [197].
As members of the S100/calgranulins protein family, S100A8 and S100A9 play crucial roles in inflammation and have been implicated in both pro-tumorigenic and anti-tumorigenic processes, depending on the cancer type [237]. CP (also referred to as S100A8/A9), a well-established biomarker of inflammation, is a heterodimeric cytosolic protein predominantly found in myeloid cells. This calcium- and a zinc-binding protein consists of S100A8 and S100A9 subunits and play a crucial role in several cellular processed, including cell cycle progression, cell survival, apoptosis, proliferation, differentiation, and migration [238]. It has been suggested that the extracellular form of CP exerts pro-inflammatory and antimicrobial effects, whereas the intracytoplasmic S100A8/A9 complex is primarily involved in cellular development, maintenance, and survival [239]. Furthermore, the extracellular form of CP modulates cellular oxidative stress and promotes inflammation-related cancer progression through the activation of key signaling pathways such as STAT3, NF-kB, and ERK-MAPK [240]. However, due to its involvement in a wide range of inflammatory processes, CP upregulation can be detected in various disease, making its specificity relatively low [241].
In a comprehensive pan-cancer analysis by Wu et al. (2022), the expression of S100A8 was found to be significantly elevated in tumor tissues [242]. Overexpression of S100A8 has been demonstrated by immunohistochemistry (IHC) in tumor tissue from various cancer types, including BC [243,244], PCa [245,246], and bladder cancer [246,247]. In OSCC, increased S100A8 expression was detected using LC-MS/MS and IF [248]. Additionally, in pancreatic ductal adenocarcinoma (PDAC), overexpression was confirmed through two-dimensional gel electrophoresis, MALDI-ToF-MS/MS, Western blot (WB), and IHC analysis [249].
A study using IHC to analyze tissue samples from bladder cancer, prostate, renal cancer, and healthy controls, CP expression was significantly elevated in bladder cancer biopsies compared to the other tumor types and healthy controls [246]. The study also revealed that urothelial cancers are associated with elevated levels of CP in urine, quantified by enzyme-linked immunosorbent assay (ELISA), further suggesting CP as a promising biomarker for the non-invasive detection of bladder cancer [246]. Using tear fluid as a non-invasive source for early BC detection, Daily et al. (2022) identified a panel of differentially expressed proteins, among which S100A8 and S100A9 were proposed as potential biomarkers due to their elevated expression in BC patients [197]. This study employed LC-MS/MS for initial protein identification, followed by ELISA validation, thereby providing strong support for the potential use of the tear proteome in BC screening and diagnosis [197].
CP has the potential to serve as diagnostic, prognostic or metastatic biomarker in several types of cancer, including those related to obstetrics and gynecology [250]. Botana-Rial et al. (2020) confirmed the value of CP as a new diagnostic biomarker for pleural effusion (PE), demonstrating with good accuracy that CP levels in benign PE (BPE) samples were higher than those in malignant pleural effusion (MPE), often associated with ovarian or BC [195].
In their study, Zhang et al. (2023) used IHC detection to show that S100A8/A9 levels were elevated in BC compared to normal tissue, with particularly high expression linked to poor differentiation, loss of hormone receptors, and HER2 positivity. Elevated S100A8/A9 levels were predictive of a worse prognosis for BC patients [243].
However, Baydar et al. (2023) found no significant difference in serum CP levels, measured by ELISA, between patients with BC requesting neo-adjuvant treatment and control groups. Interestingly, they did find the serum CP levels were associated with Ki67 level [238]. Additionally, fecal CP levels were higher in patients with early-stage BC undergoing chemotherapy, likely due to colonic- and systemic-induced inflammation and gut dysbiosis [251]. Yasar et al. (2017) found that serum S100A8 and S100A9 did not differ significantly between controls and bladder cancer patients. However, both serum and urine CP levels were significantly elevated in cancer patients compared to controls [252]. Finally, Sahin et al. (2019) reported that CP expression in tumor tissue, measured in proportion to total tissue protein level, and its urinary levels, measured by ELISA, were significantly higher in patients with high grade, muscle-invasive primary bladder cancer [253].
In the previously cited study by Botana-Rial et al. (2020), CP levels in pleural fluid were assessed to differentiate between benign and malignant pleural effusion [195]. The study found a sensitivity of 96% and specificity of 60% for a cut-off value of ≤6233.2 ng/mL [195]. A meta-analysis by Ye et al. (2018) reported that fecal CP had a pooled sensitivity of 83% and specificity of 61% for CRC detection [254]. A strong correlation was observed between procalcitonin (PCT), the prohormone of calcitonin (CT), and CT levels in patients with medullary thyroid carcinoma (MTC), PCT emerging as a complementary MTC tumor biomarker [255]. While fecal CP demonstrates promise as a biomarker for certain gastrointestinal cancers, its sensitivity and specificity vary across studies and populations. Factors such as disease stage, assay methods, and patient’s history can influence these values. CP’s role in LB for cancer detection remains under investigation.
Although the individual plasma biomarkers had some ability to predict cancer, the combination panel of more biomarkers have good predictive value for cancer. For example, a study by Sun et al. (2020) showed that CEA, osteopontin (OPN), and Dickkopf 1 (DKK1) protein may be involved in occurrence, progression, and migration of non-small cell lung cancer (NSCLC) [256]. The study revealed that the serum CEA sensitivity and specificity in NSCLC detection were 86.25% and 70%, respectively; the sensitivity and specificity of serum OPN were 71.61% and 91,25%, respectively; the sensitivity and specificity of serum DKK1 were 92.5% and 65%, respectively; the sensitivity and specificity of further combination of serum CEA and OPN were 87.5% and 86.67%, respectively; the sensitivity and specificity of the combination of serum CEA and DKK1 in the diagnosis of NSCLC were 92.5% and 76.67%, respectively [256]. These authors concluded that these three methods can be used as biological markers for the diagnosis of NSCLC [256]. A panel called the four-protein biomarker panel (4MP), including pro-SFPB, CA125, CYFRA21-1, and CEA, has been validated for assessment of lung cancer risk [257]. This panel, has a sensitivity of 63% and a specificity of 83% [258]. Additionally, a study by Nolen et al. (2015) revealed that urine biomarkers may provide screening and diagnostic properties which exceed those reported for serum biomarkers and are necessary for clinical development [259].

5. Evaluation of Diagnostic Performance in Cancer Detection for Clinical Applications

An analysis of the sensitivity and specificity values for several commonly used cancer detection methods allows for a comparative assessment, leading to several important considerations regarding the reliability, effectiveness, and clinical applicability of these diagnostic approaches (Table 3). Fine needle aspiration biopsy/cytology (FNAB/FNAC) and core needle biopsy (CBB) stand out with high sensitivity (74 or 97%) and high specificity (96% and 98%, respectively) for BC detection [67,68,69]. These methods are highly reliable in detecting BC, particularly with the highest accuracy reported for CNB. 18F-fluorodeoxyglucose Positron Emission Tomography/Computed Tomography (FDG-PET/CT) shows impressive sensitivity (97.4%) and specificity (91.2%), making it effective for detecting distant metastasis in BC [99]. Thus, the specificity and sensitivity of FDG-PET/CT were significantly higher than those of conventional imaging (CT, US, radiography, and skeletal scintigraphy), with 85.9% sensitivity and 67.3% specificity for detection of distant BC metastases [99]. Modern film mammography shows high sensitivity (71.2% for biennial screening) and good specificity (92.6%) for asymptomatic Japanese women aged 40-49 years [90]. Digital mamography demonstrate high sensitivity (97%) but moderate specificity (64.5%), indicating its utility in early detection but challenges in differentiating malignant from benign cases without further diagnostic procedures [87,88]. mpMRI (multi-parametric MRI), with a sensitivity of 93%, is a valuable tool for PCa detection, although its specificity is lower (48%) [142]. Concluding here, FNAB/FNAC, FDG-PET/CT, mammography, and mpMRI are considered to have high sensitivity in cancer detection. HE4 has demonstrated the highest specificity (97.87%) in diagnosis of epithelial ovarian cancer (EOC), making it reliable in distinguish malignant from benign cases, particularly in later stages [43]. Transrectal ultrasonography (TRUS)-guided biopsy and human epididymis protein has specificity as a strength in diagnostic methods. Thus, TRUS-guided biopsy shows high specificity (96%) for PCa detection and has moderate sensitivity (48%) [142].
Among the cancer detection methods characterized by moderate to low sensitivity, several examples can be highlighted. BSE, although simple and non-invasive, shows low sensitivity (20-30%) but relatively high specificity (87.4%). While it is not reliable for primary BC detection, it can still serve as a complementary technique alongside more advanced methods [83,260]. Serum-PSA testing, with sensitivity ranging from 21% to 32% depending on the threshold, is similarly limited. It is particularly unreliable for detecting PCa at lower PSA levels, as it often fails to identify early-stage disease [142]. In CRC screening, fecal CP at a CR50 µg/g cutoff offers moderate sensitivity (83%) but lower specificity (61%), making it a useful, though not definitive, screening tool [261]. Additionally, CP has demonstrated high sensitivity (96%) in distinguishing between benign and malignant pleural effusions, suggesting promising potential for non-invasive lung diagnosis [195].
Serum CEA levels used in detection of non-small cell lung cancer (NSCLC) exhibit relatively low specificity (70%) despite a sensitivity of 86.25%. This limitation is partly due to CEA elevation in various other cancer (such as breast, prostate, and gastric cancer), as well as in non-cancerous conditions like cirrhosis and COPD [256]. These findings highlight the importance of employing multi-biomarker panels to increase diagnostic accuracy and minimize false positive results. For instance, the combination of serum CEA and OPN demonstrates high sensitivity (87.5%) and improved specificity (86.67%), showing potential for more accurate detection and differentiation of lung and other cancers [256]. Similarly, pairing serum CEA with DKK1 yields an even higher combined sensitivity (92.5%) and moderate specificity (76.67%), representing a promising diagnostic approach for malignancies such as non-small cell lung cancer (NSCLC) [256]. The combination of matrix metalloproteinase 9 (MMP9) and KPYM (pyruvate kinase M1/2) exhibited 94% sensitivity and 87% specificity for detecting endometrial cancer (EC), while the combination catenin beta 1 (CTNB1), exportin 2 (XPO2), and macrophage-capping protein (CAPG) achieved 95% sensitivity and 96% specificity for the discrimination of endometrioid EC and serous EC [262]. These biomarkers were measured in the fluid fraction of uterine aspirates by liquid chromatography-parallel reaction monitoring (LC-PRM) and have been analyzed using the latest generation of targeted-MS aquisition [262].
The development of multi-protein biomarkers panels that combine high-sensitivity and high-specificity markers is crucial for enhancing diagnostic accuracy and minimizing misdiagnosis. LB-based strategies are increasingly recognized for their potential in non-invasive, real-time monitoring of cancer progression and evaluation of therapeutic response, although further research id required to improve their specificity. The integration of AI and ML in the analysis of large datasets generated from diverse detection methods may help overcome current linitations by anabling more personalized and precise diagnoses. Given the wide variability in sensitivity and specificity among existing cancer detection techniques—some offering high sensitivity at the expense of specificity, and vice versa—a multimodal, multi-biomarker approach, supported by AI and ML advancements, presents a promising path toward more accurate, reliable, and rapid cancer detection.
The integration of conventional cancer detection methods—such as imaging techniques, tissue biopsies, and serum-based markers—with proteomics-based LB offers a powerful, complementary approach to cancer diagnosis and monitoring. While conventional methods remain the cornerstone for initial detection and staging, they often lack sensitivity for early-stage disease or are invasive in nature. Proteomics-based LB, by analyzing protein signatures from blood or other body fluids, provides a minimally invasive tool capable of capturing dynamic changes in the tumor environment. When combined, these approaches can enhance diagnostic accuracy, enable earlier detection, and allow for more precise tracking of disease progression and treatment response. This synergy not only improves patient outcomes but also supports the move toward more personalized oncology care.
Table 3. Comparative sensitivity and specificity of different methods used in cancer detection.
Table 3. Comparative sensitivity and specificity of different methods used in cancer detection.
Detection MethodsSensitivitySpecificityClinical Applications
BSE20% to 30%; 58,3%87.4%BC detection [83,260]
iBE34.3% to 86%,59% to 94%BC detection [85]
CBE40% to 69%-BC detection [263]
CBE (average-risk women)-99.4% (screening); 68.7% (diagnostic)[264]
CBE (increased-risk women)-97.1% (screening)
57.1% (doagnostic)
DRE51%59%PCa detection [141]
serum PSA32% (3.1 ng/mL);
21% (4 ng/mL)
-PCa detection [142]
US 80.1% (overall pooled sensitivity)88.4%BC diagnosis [87]
US 89.2% (in low and middle-income country)99.1%
mammography77–95%-BC detection [263]
modern film mammography alone, without CBE, among asymptomatic Japanese women (40-49 years)47.4%-BC screening [90]
biennial modern film mammography, without CBE, among asymptomatic Japanese women (40-49 years)71.7%92.6%
digital mammography97%64.5%,
(89.3% accuracy)
BC detection [87,88]
FNAB/FNAC74%/97%96% (95% accuracy)BC detection [67,68,69]
CNB95%/97%98% (96% accuracy)BC detection [67,68,69]
conventional imaging (CT, US, radiography, skeletal scintigraphy)85.9%67.3%detection of BC distant metastasis [99]
salivary tests71.7%72.7%BC detection [168]
salivary chemerin and MMP-9, and serum chemerin100%100%differentiating OSCC from OPMLs [176]
CEUS (polled sensitivity)70%74%PCa detection [265]
grey-scale TRUS40% to 50%40% to 50%
TRUS-guided biopsy48%96%PCa detection [142]
mpMRI93%48%
mpMRI and PI-RADS89%73%PCa detection [142,266]
MRI72%96% PCa staging [142]
FDG-PET/CT97.4%91.2%detection of BC distant metastases [99]
latest-generation digital PET/CT96.4%86.4%detection of cervical LN metastases in HNSCC [100]
HE476% to 88% (depending on disease stage)97.87%OC detection [43]
4MP (pro-SFPB, CA125, CYFRA21-1, CEA)63%83%distinguishing benign from malign indeterminate pulmonary nodules [258]
serum CEA86.25%70%NSCLC diagnosis [256]
serum OPN71.61%91.25%
serum DKK192.5%65%
serum CEA and OPN87.5%86.67%
serum CEA and DKK192.5%76.67%
MMP9 and KPYM (uterine aspirate)94%87%EC detection [262]
CTNB1, XPO2, and CAPG (uterine aspirate)95%96%discrimination of endometrioid EC and serous EC [262]
blood test based on EVs71.2%99.5%detection of OC, PC, bllader cancer (early-stage) [16]
calprotectin (CP)
(≤6233.2 ng/mL cutoff)
96%60%benign and malignant pleural effusion differentiation [195]
fecal CP
(50 µg/g cutoff)
83%61%CRC detection [254]
76.9%88% esofago-gastric cancer detection [261]
MasSpec Pen technology95.5%89.7%differentiation between normal pancreas and PC [226]
92%96.2%differentiation between bile duct and pancreatic cancer [226]
96.4%96.2%prediction of cancer subtype of lung and thyroid cancer [63]
Abbreviations: BC—breast cancer; BSE—breast self-examination; CA19-9—carbohydrate antigen 19-9; CAPG—macrophage-capping protein; CBE—clinical breast examination; CEA—carcinoma embryonic antigen; CEUS—contrast-enhanced ultrasound; CNB—core needle biopsy; COPD—chronic obstructive pulmonary disease; CRC—colorectal cancer; CT—computed tomography; DKK1—Dickkopf-1; DRE—digital rectal examination; EC—endometrial cancer; EVs—extracellular vesicles; FDG-PET/CT—18F-fluorodeoxyglucose Positron Emission Tomography/Computed Tomography; FNAB/FNAC—fine needle aspiration biopsy/cytology; 4M—four-protein biomarker panel; GC—gastric cancer; HE4—human epididymis protein 4; HNSCC—head and neck squamous cell carcinoma; iBE—Intelligent Breast Exam; KPYM—pyruvate kinase M1/2; LN—lymph node; MMP9—matrix metalloproteinase 9; mpMRI—multi-parametric MRI; MRI—magnetic resonance imaging; NSCLC—non-small cell lung cancer; OC—ovarian cancer; OPMLs—oral premalignant lesions; OSP—osteopontin; PC—pancreatic cancer; PCa-prostate cancer; PI-RADS—Prostate Imaging Reporting and Data System; PSA—prostate-specific antigen; SCLC—small cell lung cancer; TRUS—transrectal ultrasound; US—ultrasonography; XPO2—exportin 2.

6. Future Perspectives

Despite significant advances in cancer diagnosis, the search for ideal methods to detect malignant tumor remains ongoing. Researchers, clinicians, and even patients, particularly from the perspective of participatory medicine, continue to seek more reliable tools and solutions [1]. Early cancer detection, with minimal discomfort, enables patients to begin personalized therapy sooner, thereby improving the likelihood of a successful outcome and reducing the physical and physiological impact of invasive procedures.
However, all current methods used in cancer screening and diagnosis have notable limitations. Conventional approaches often suffer from reduced sensitivity, specificity, and accuracy. They are often invasive, limited to certain types of cancer, and typically detect one or few cancer biomarkers. Furthermore, they methods struggle to monitor the dynamic development of tumors or capture the full molecular landscape within tumors and cancerous cells. In addition, some conventional methods carry inherent risk of complications, are time-consuming, labor-intensive, and resources-consuming, and may be limited in accessibility due to high costs. Consequently, these limitations often lead to late cancer detection, overdiagnosis, overtreatment, unnecessary surgery, or delayed treatment decisions. In some cases, results are also subject to interpretation variability and human error.
This review examines the evolution of cancer detection methods, highlighting the transition from conventional techniques to emerging innovation such as liquid biopsy (LB) and molecular biomarkers. It emphasizes the need for enhanced accuracy, reduced invasiveness, and early cancer detection, as well as the facilitation of personalized treatment strategies.
A comprehensive overview is provided on various cutting-edge approaches, including multimodal imaging, genomic testing, and pan-cancer analysis, in conjunction with multi-omics strategies, which offer a more holistic understanding of cancer biology. The review also explores the growing role of AI and ML in improving diagnostic accuracy and predicting treatment response. Additionally, we highlighted the potential of proteome-based plasma tests and LB, including the sensitive detection of CTCs, which enable non-invasive monitoring of cancer progression and treatment efficacy. The use of extracellular vesicles (EVs) in protein-based blood tests represents another promising avenue, offering insights into tumor dynamics, molecular signatures, and intra-tumoral heterogeneity.
Furthermore, single-cell mass spectrometry analysis of multiplex proteomes and microscopy-based functional single-cell proteomic profiling provide in-depth examination of tumor intraheterogeneity and the dysregulated pathways that drive carcinogenesis and tumor development. We also discuss the development of MSI and MasSpec Pen technologies, which can revolutionize cancer detection by offering rapid, accurate, and accessible testing outside conventional clinical settings.
The development of PoC diagnostic tool is accelerating the speed, ease, cost-effectiveness, and on-site availability in cancer testing, thanks to advances in biosensor technologies powered by innovative nanomaterials [230].
Today, nanotechnologies enable real-time detection of tumor biomarkers, paving the way for early diagnosis, personalized treatment plans, and continuous cancer monitoring. As the design and functionality of nanomaterials continuous to evolve, improvements in biosensor accuracy, sensing performance, specificity, and versatility will greatly enhance the quality of life for cancer patients. Recent progress in LB using optical nanomaterials offers the opportunity to discover novel biomarkers and deepen our understanding of cancer-related mechanisms. LB protein biomarker profiling could be integrated into routine health monitoring through home-based LB collection kits.
Together, these advancements are creating a pathway for more personalized, precise cancer detection and management. As these technologies evolve, they hold immense potential to enhance early detection, improve diagnostic accuracy, and optimize treatment strategies. Ultimately, these innovations offer hope for more effective and less invasive methods to fight against cancer.
Over next decades, LB is likely to evolve into a routine, highly precise, and universally accessible tool that redefines not only cancer detection, but also overall health monitoring.
The future of LB may include:
  • Ultra-early cancer detection, on even cellular or molecular level, identifying malignant transformation of cells and signaling pathways before they become anatomically detectable;
  • Wearable or implantable biosensors continuously sampling and analyzing blood or interstitial fluids in real-time, alerting both patient and physician to molecular changes before symptoms appear;
  • Fully personalized protein biomarker panels generated through ML and AI, dynamically adapting to individual genomic and proteomic profiles, lifestyle factors, and environmental exposures;
  • PoC diagnostics in everyday life, including smartwatches or home health stations, enabling regular health checks without needing clinical visits;
  • Integration with preventive medicine, where LB not only detects cancer but predicts risk, monitors predisposition, and guides proactive interventions years before clinical cancer manifests.
In this feature, the role of tissue biopsy may become limited to rare or complex cases, while LB becomes the foundation of longitudinal health tracking, combining convenience with unprecedented depth of biological insights.
Going further, LB could become an inseparable part of routine life, converging to a real-time, continuous monitoring of the body molecular and cellular health, where nanobots circulate in the bloodstream and collect and transmits real-time data on cancer biomarkers, disease progression, and overall health, allowing for immediate detection of even slightest abnormalities, far before symptoms emerge. These nanobots will also be equipped to adjust dysregulated signaling pathways. Advanced LB systems will be able to detect at early stage not just cancer, but also for a wide range of diseases. Cancer will be detected on a molecular level. Consequently, human lifespan may see a significant increase sue to the continuous monitoring of biomarkers, allowing for precise management of aging and prevention of age-related diseases, including cancer. Last, but not least, medical care will be more preventive and predictive, and patients will be deeply involved in participatory medicine. Furthermore, it will be an era of preemptive medicine, that refers to an advances approach in healthcare, where the focus is on preventing cancer before its even occurs rather just treating it once it manifests.

7. Conclusions

Cancer detection has evolved significantly. Transitioning from conventional methods to innovative, non- or minimally invasive approaches aims to enhance early diagnosis, precision, and treatment outcomes. This review highlights current and emerging diagnostic technologies, including LB, molecular biomarkers, and AI.LB, utilizing biomarkers in bodily fluids. It offers a method for monitoring cancer progression, detecting tumors at molecular levels, and predicting treatment responses. Despite its promising potential, the clinical application of LB faces several challenges. This includes issues related to sensitivity and specificity, as low levels of tumor biomarkers in early-stage cancers can lead to false negatives, while non-cancerous conditions may produce false positives. Additionally, the heterogeneity of tumors complicates the detection of all cancer types, and the lack of standardized protocols for sample collection, processing, and analysis creates variability in results. Technological limitations in detecting low concentrations of biomarkers and the high cost of advanced equipment also pose significant barriers to widespread adoption. Furthermore, the interpretation of LB results remains complex, requiring clear clinical guidelines and established threshold levels for reliable decision-making. Ethical and privacy concerns regarding patients’ data, alongside the need for large-scale validation and clinical integration, hinder the universal application of LB. While LB holds great promise for revolutionizing cancer detection, overcoming these challenges is critical to ensuring its accuracy, accessibility, and integration into routine clinical practice.
Proteoforms, the diverse variants of proteins arising from genetic and PTMs, play a crucial role in the molecular landscape of cancer. Their specific expression patterns often reflect tumor presence, subtype, and progression more accurately than conventional biomarkers. Detecting these proteoforms, especially through minimally invasive LBs, offers a promising avenue for early cancer diagnosis when intervention can be most effective. By capturing the complexity and heterogeneity of tumors at the proteoform level, clinicians can improve diagnostic precision, tailor treatments to individual patients, and ultimately enhance clinical outcomes. Ongoing innovations in proteoform detection technologies are therefore essential to realizing the full potential of personalized oncology.
Continued advancements in technology, standardization, and bioinformatics, along with ongoing research into the clinical significance of LB biomarkers, are essential for realizing the full potential of LB in cancer care. The integration of AI and ML further enhances diagnostic accuracy by enabling rapid data analysis from diverse sources, including genomics, proteomics, and imaging. Additionally, advancements in nanotechnology, single-cell analysis, and mass spectrometry are reshaping the landscape of cancer detection, enabling high-sensitivity, real-time monitoring of biomarkers. Despite the progress, challenges such as limited specificity, high costs, and the need for multi-biomarker panels remain. The future of cancer diagnostics is poised to focus on personalized, real-time detection using wearable biosensors, lab-on-a-chip technologies, and advanced LB systems, promising enhanced early detection, reduced invasiveness, and improved patient outcomes. In the long term, these innovations may redefine cancer detection and treatment, leading to a more proactive, predictive, and patient-centered healthcare approach.

Author Contributions

Conceptualization, A.-N.N. and C.C.D.; literature search, A.-N.N., P.S.B., C.-L.J., N.W., H.M., V.T.N. and C.C.D.; writing—original draft preparation, A.-N.N., P.S.B., C.-L.J., N.W., H.M., V.T.N. and C.C.D.; writing—review and editing A.-N.N., P.S.B., C.-L.J., N.W., H.M., V.T.N. and C.C.D.; project administration, C.C.D.; funding acquisition, C.C.D. All authors have read and agreed to the published version of the manuscript.

Funding

This publication was supported in part by the National Cancer Institute of the National Institutes of Health under Award Number R15CA260126. The content is solely the responsibility of the authors and does not necessarily represent the official views of the National Institutes of Health. P.S.B., V.T.N., and C.C.D. were in part supported through the Erasmus+ exchange program, granted to “Al. I. Cuza” University of Iasi, Romania.

Acknowledgments

The authors thank the members of the Biochemistry & Proteomics Laboratories for the pleasant working environment.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Siffels, L.E.; Sharon, T.; Hoffman, A.S. The participatory turn in health and medicine: The rise of the civic and the need to ‘give back’ in data-intensive medical research. Humanit. Soc. Sci. Commun. 2021, 8, 306. [Google Scholar] [CrossRef]
  2. Loud, J.T.; Murphy, J. Cancer Screening and Early Detection in the 21st Century. Semin. Oncol. Nurs. 2017, 33, 121–128. [Google Scholar] [CrossRef]
  3. Ginghina, O.; Hudita, A.; Zamfir, M.; Spanu, A.; Mardare, M.; Bondoc, I.; Buburuzan, L.; Georgescu, S.E.; Costache, M.; Negrei, C.; et al. Liquid Biopsy and Artificial Intelligence as Tools to Detect Signatures of Colorectal Malignancies: A Modern Approach in Patient’s Stratification. Front. Oncol. 2022, 12, 856575. [Google Scholar] [CrossRef]
  4. Mukherjee, N.; Chatterjee, N.; Manna, K.; Das Saha, K. Chapter 2-Types of cancer diagnostics, the current achievements, and challenges. In Biosensor Based Advanced Cancer Diagnostics; Khan, R., Parihar, A., Sanghi, S.K., Eds.; Academic Press: Cambridge, UK, 2022; pp. 27–45. [Google Scholar] [CrossRef]
  5. Chubak, J.; Burnett-Hartman, A.N.; Barlow, W.E.; Corley, D.A.; Croswell, J.M.; Neslund-Dudas, C.; Vachani, A.; Silver, M.I.; Tiro, J.A.; Kamineni, A. Estimating Cancer Screening Sensitivity and Specificity Using Healthcare Utilization Data: Defining the Accuracy Assessment Interval. Cancer Epidemiol. Biomark. Prev. 2022, 31, 1517–1520. [Google Scholar] [CrossRef] [PubMed]
  6. Trevethan, R. Sensitivity, Specificity, and Predictive Values: Foundations, Pliabilities, and Pitfalls in Research and Practice. Front. Public Health 2017, 5, 307. [Google Scholar] [CrossRef]
  7. Santini, A.; Man, A.; Voidăzan, S. Accuracy of Diagnostic Tests. J. Crit. Care Med. 2021, 7, 241–248. [Google Scholar] [CrossRef] [PubMed]
  8. McKay, K.M.; Lim, L.L.; Van Gelder, R.N. Rational laboratory testing in uveitis: A Bayesian analysis. Surv. Ophthalmol. 2021, 66, 802–825. [Google Scholar] [CrossRef] [PubMed]
  9. Lipscomb, J.; Horton, S.; Kuo, A.; Tomasetti, C. Evaluating the impact of multicancer early detection testing on health and economic outcomes: Toward a decision modeling strategy. Cancer 2022, 128, 892–908. [Google Scholar] [CrossRef]
  10. Englisz, A.; Smycz-Kubańska, M.; Mielczarek-Palacz, A. Sensitivity and Specificity of Selected Biomarkers and Their Combinations in the Diagnosis of Ovarian Cancer. Diagnostics 2024, 14, 949. [Google Scholar] [CrossRef]
  11. Combes, G.F.; Vučković, A.-M.; Bakulić, M.P.; Antoine, R.; Bonačić-Koutecky, V.; Trajković, K. Nanotechnology in Tumor Biomarker Detection: The Potential of Liganded Nanoclusters as Nonlinear Optical Contrast Agents for Molecular Diagnostics of Cancer. Cancers 2021, 13, 4206. [Google Scholar] [CrossRef]
  12. Forgrave, L.M.; Wang, M.; Yang, D.; DeMarco, M.L. Proteoforms and their expanding role in laboratory medicine. Pract. Lab. Med. 2022, 28, e00260. [Google Scholar] [CrossRef]
  13. Michela, B. Liquid Biopsy: A Family of Possible Diagnostic Tools. Diagnostics 2021, 11, 1391. [Google Scholar] [CrossRef] [PubMed]
  14. Ma, L.; Guo, H.; Zhao, Y.; Liu, Z.; Wang, C.; Bu, J.; Sun, T.; Wei, J. Liquid biopsy in cancer: Current status, challenges and future prospects. Signal Transduct. Target. Ther. 2024, 9, 336. [Google Scholar] [CrossRef] [PubMed]
  15. Deng, Z.; Wu, S.; Wang, Y.; Shi, D. Circulating tumor cell isolation for cancer diagnosis and prognosis. EBioMedicine 2022, 83, 104237. [Google Scholar] [CrossRef]
  16. Hinestrosa, J.P.; Kurzrock, R.; Lewis, J.M.; Schork, N.J.; Schroeder, G.; Kamat, A.M.; Lowy, A.M.; Eskander, R.N.; Perrera, O.; Searson, D.; et al. Early-stage multi-cancer detection using an extracellular vesicle protein-based blood test. Commun. Med. 2022, 2, 29. [Google Scholar] [CrossRef]
  17. Giunta, E.F.; Malapelle, U.; Russo, A.; De Giorgi, U. Blood-based liquid biopsy in advanced prostate cancer. Crit. Rev. Oncol. Hematol. 2024, 194, 104241. [Google Scholar] [CrossRef]
  18. Kong, T.; Qu, Y.; Zhao, T.; Niu, Z.; Lv, X.; Wang, Y.; Ding, Q.; Wei, P.; Fu, J.; Wang, L.; et al. Identification of novel protein biomarkers from the blood and urine for the early diagnosis of bladder cancer via proximity extension analysis. J. Transl. Med. 2024, 22, 314. [Google Scholar] [CrossRef] [PubMed]
  19. Montero-Calle, A.; Garranzo-Asensio, M.; Torrente-Rodríguez, R.M.; Montiel, V.R.-V.; Poves, C.; Dziaková, J.; Sanz, R.; del Arco, C.D.; Pingarrón, J.M.; Fernández-Aceñero, M.J.; et al. p53 and p63 Proteoforms Derived from Alternative Splicing Possess Differential Seroreactivity in Colorectal Cancer with Distinct Diagnostic Ability from the Canonical Proteins. Cancers 2023, 15, 2102. [Google Scholar] [CrossRef]
  20. Montero-Calle, A.; Garranzo-Asensio, M.; Moreno-Casbas, M.T.; Campuzano, S.; Barderas, R. Autoantibodies in cancer: A systematic review of their clinical role in the most prevalent cancers. Front. Immunol. 2024, 15, 1455602. [Google Scholar] [CrossRef]
  21. Preethi, K.; Jeyarajan Selvakumaran, S.C.; Ross, K.; Jayaraman, S.; Deusdedit, T.; Sekar, D. Liquid biopsy: Exosomal microRNAs as novel diagnostic and prognostic biomarkers in cancer. Mol. Cancer 2022, 21, 54. [Google Scholar] [CrossRef]
  22. van der Poort, E.K.J.; van Ravesteyn, N.T.; van den Broek, J.J.; de Koning, H.J. The Early Detection of Breast Cancer Using Liquid Biopsies: Model Estimates of the Benefits, Harms, and Costs. Cancers 2022, 14, 2951. [Google Scholar] [CrossRef] [PubMed]
  23. Manea, I.; Iacob, R.; Iacob, S.; Cerban, R.; Dima, S.; Oniscu, G.; Popescu, I.; Gheorghe, L. Liquid biopsy for early detection of hepatocellular carcinoma. Front. Med. 2023, 10, 1218705. [Google Scholar] [CrossRef] [PubMed]
  24. Zhu, Y.; Zhang, H.; Chen, N.; Hao, J.; Jin, H.; Ma, X. Diagnostic value of various liquid biopsy methods for pancreatic cancer: A systematic review and meta-analysis. Medicine 2020, 99, e18581. [Google Scholar] [CrossRef]
  25. Li, Y.-Z.; Kong, S.-N.; Liu, Y.-P.; Yang, Y.; Zhang, H.-M. Can Liquid Biopsy Based on ctDNA/cfDNA Replace Tissue Biopsy for the Precision Treatment of EGFR-Mutated NSCLC? J. Clin. Med. 2023, 12, 1438. [Google Scholar] [CrossRef]
  26. Bogdan, B.; Hossein, A.; Mohammad, H.F.; Ashkan, A. Novel proteomics-based plasma test for early detection of multiple cancers in the general population. BMJ Oncol. 2024, 3, e000073. [Google Scholar] [CrossRef]
  27. Tessitore, A.; Gaggiano, A.; Cicciarelli, G.; Verzella, D.; Capece, D.; Fischietti, M.; Zazzeroni, F.; Alesse, E. Serum Biomarkers Identification by Mass Spectrometry in High-Mortality Tumors. Int. J. Proteom. 2013, 2013, 125858. [Google Scholar] [CrossRef]
  28. Merriel, S.W.D.; Pocock, L.; Gilbert, E.; Creavin, S.; Walter, F.M.; Spencer, A.; Hamilton, W. Systematic review and meta-analysis of the diagnostic accuracy of prostate-specific antigen (PSA) for the detection of prostate cancer in symptomatic patients. BMC Med. 2022, 20, 54. [Google Scholar] [CrossRef]
  29. De Souza Dutra, C.; Da Cruz Schafhauser, D.; Hentz, M.; Mayer, N.R.; Pinheiro, R.M.; Baierle, G.; Kist, D.R.; Bullé, D.J.; Donaduzzi, R.C.; Bohmgahren, M.F.; et al. Urinary endogenous peptides as biomarkers for prostate cancer. Oncol. Lett. 2023, 25, 173. [Google Scholar] [CrossRef] [PubMed]
  30. 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]
  31. Saxby, H.; Mikropoulos, C.; Boussios, S. An Update on the Prognostic and Predictive Serum Biomarkers in Metastatic Prostate Cancer. Diagnostics 2020, 10, 549. [Google Scholar] [CrossRef]
  32. Nepal, A.; Sharma, P.; Bhattarai, S.; Mahajan, Z.; Sharma, A.; Sapkota, A.; Sharma, A. Extremely Elevated Prostate-Specific Antigen in Acute Prostatitis: A Case Report. Cureus 2023, 15, e43730. [Google Scholar] [CrossRef]
  33. Khoo, A.; Liu, L.Y.; Nyalwidhe, J.O.; Semmes, O.J.; Vesprini, D.; Downes, M.R.; Boutros, P.C.; Liu, S.K.; Kislinger, T. Proteomic discovery of non-invasive biomarkers of localized prostate cancer using mass spectrometry. Nat. Rev. Urol. 2021, 18, 707–724. [Google Scholar] [CrossRef] [PubMed]
  34. Loeb, S.; Catalona, W.J. The Prostate Health Index: A new test for the detection of prostate cancer. Ther. Adv. Urol. 2014, 6, 74–77. [Google Scholar] [CrossRef] [PubMed]
  35. Chiu, P.K.-F.; Liu, A.Q.; Lau, S.-Y.; Teoh, J.Y.-C.; Ho, C.-C.; Yee, C.-H.; Hou, S.-M.; Chan, C.-K.; Tang, W.-L.; Bangma, C.H.; et al. A 2-year prospective evaluation of the Prostate Health Index in guiding biopsy decisions in a large cohort. BJU Int. 2025, 135, 71–77. [Google Scholar] [CrossRef] [PubMed]
  36. Punnen, S.; Pavan, N.; Parekh, D. Finding the Wolf in Sheep’s Clothing: The 4Kscore Is a Novel Blood Test That Can Accurately Identify the Risk of Aggressive Prostate Cancer. Rev. Urol. 2015, 17, 3–13. [Google Scholar]
  37. Cui, Y.; Cao, W.; Li, Q.; Shen, H.; Liu, C.; Deng, J.; Xu, J.; Shao, Q. Evaluation of prostate cancer antigen 3 for detecting prostate cancer: A systematic review and meta-analysis. Sci. Rep. 2016, 6, 25776. [Google Scholar] [CrossRef]
  38. He, Y.; Lu, H.; Zhang, L. Chapter Ten-Serum AFP levels in patients suffering from 47 different types of cancers and noncancer diseases. In Progress in Molecular Biology and Translational Science; Zhang, L., Ed.; Academic Press: Cambridge, UK, 2019; Volume 162, pp. 199–212. [Google Scholar]
  39. Miyoshi, A.; Takashi, M.; Komiya, S.; Mimura, M.; Nagamatsu, M.; Yokoi, T. Highly Elevated Level of Serum CA125 Produced by a Large Uterine Leiomyoma in a 20-Year-Old Woman. J. Clin. Gynecol. Obstet. 2015, 4, 275–278. [Google Scholar] [CrossRef]
  40. Karimi-Zarchi, M.; Dehshiri-Zadeh, N.; Sekhavat, L.; Nosouhi, F. Correlation of CA-125 serum level and clinico-pathological characteristic of patients with endometriosis. Int. J. Reprod. Biomed. 2016, 14, 713–718. [Google Scholar] [CrossRef] [PubMed]
  41. Khadoor, S.; Ibrahim, R.; Redwan, F. The Significance of Serum CA-125 in patients with Polycystic Ovarian Syndrome and its Association with their Hormonal Status. Res. J. Pharm. Technol. 2024, 17, 5445–5451. [Google Scholar] [CrossRef]
  42. Kim, J.H.; Park, B.R.; Yang, W.J. Dilution effect of serum CA125 and CA19-9 over a cutoff value, according to obesity. Int. J. Biol. Markers 2015, 30, 122–126. [Google Scholar] [CrossRef]
  43. Li, Y.; Wang, Z.-C.; Luo, L.; Mu, C.-Y.; Xu, J.; Feng, Q.; Li, S.-B.; Gu, B.; Ma, P.; Lan, T. The clinical value of the combined detection of sEGFR, CA125 and HE4 for epithelial ovarian cancer diagnosis. Eur. Rev. Med. Pharmacol. Sci. 2020, 24, 604–610. [Google Scholar] [PubMed]
  44. Sun, S.; Wei, L.; Zou, L.; Wang, T.; Liu, Z.; He, J.; Sun, X.; Zhong, W.; Zhao, F.; Li, X.; et al. Preoperative serum CA125 level and age at diagnosis: An effective prognosis prediction tool for patients with early-stage endometrial cancer. Asia-Pac. J. Clin. Oncol. 2023, 19, e258–e266. [Google Scholar] [CrossRef]
  45. Hao, C.; Zhang, G.; Zhang, L. Chapter Eleven-Serum CEA levels in 49 different types of cancer and noncancer diseases. In Progress in Molecular Biology and Translational Science; Zhang, L., Ed.; Academic Press: Cambridge, UK, 2019; Volume 162, pp. 213–227. [Google Scholar]
  46. Blatt, S.; Kämmerer, P.W.; Krüger, M.; Surabattula, R.; Thiem, D.G.E.; Dillon, S.T.; Al-Nawas, B.; Libermann, T.A.; Schuppan, D. High-Multiplex Aptamer-Based Serum Proteomics to Identify Candidate Serum Biomarkers of Oral Squamous Cell Carcinoma. Cancers 2023, 15, 2071. [Google Scholar] [CrossRef] [PubMed]
  47. Ding, Z.; Wang, N.; Ji, N.; Chen, Z.-S. Proteomics technologies for cancer liquid biopsies. Mol. Cancer 2022, 21, 53. [Google Scholar] [CrossRef]
  48. Roy, D.; Pascher, A.; Juratli, M.A.; Sporn, J.C. The Potential of Aptamer-Mediated Liquid Biopsy for Early Detection of Cancer. Int. J. Mol. Sci. 2021, 22, 5601. [Google Scholar] [CrossRef]
  49. Wu, L.; Niu, Q.; Yang, C. Aptamer Molecular Evolution for Liquid Biopsy. In Handbook of Chemical Biology of Nucleic Acids; Sugimoto, N., Ed.; Springer Nature Singapore: Singapore, 2023; pp. 1453–1496. [Google Scholar] [CrossRef]
  50. Lin, B.; Jiang, J.; Zhou, X. Recent advances in design strategies of aptamer-based liquid biopsy. J. Polym. Sci. 2024, 62, 2848–2870. [Google Scholar] [CrossRef]
  51. de Araújo, N.S.; Moreira, A.d.S.; Abreu, R.d.S.; Junior, V.V.; Antunes, D.; Mendonça, J.B.; Sassaro, T.F.; Jurberg, A.D.; Ferreira-Reis, R.; Bastos, N.C.; et al. Aptamer-Based Recognition of Breast Tumor Cells: A New Era for Breast Cancer Diagnosis. Int. J. Mol. Sci. 2024, 25, 840. [Google Scholar] [CrossRef] [PubMed]
  52. Sun, D.; Ma, Y.; Wu, M.; Chen, Z.; Zhang, L.; Lu, J. Recent progress in aptamer-based microfluidics for the detection of circulating tumor cells and extracellular vesicles. J. Pharm. Anal. 2023, 13, 340–354. [Google Scholar] [CrossRef] [PubMed]
  53. Hanžek, A.; Ducongé, F.; Siatka, C.; Duc, A.-C.E. Identification and Characterization of Aptamers Targeting Ovarian Cancer Biomarker Human Epididymis Protein 4 for the Application in Urine. Cancers 2023, 15, 452. [Google Scholar] [CrossRef]
  54. Yoon, S.; Rossi, J.J. Emerging cancer-specific therapeutic aptamers. Curr. Opin. Oncol. 2017, 29, 366–374. [Google Scholar] [CrossRef]
  55. Mahmoudian, F.; Ahmari, A.; Shabani, S.; Sadeghi, B.; Fahimirad, S.; Fattahi, F. Aptamers as an approach to targeted cancer therapy. Cancer Cell Int. 2024, 24, 108. [Google Scholar] [CrossRef] [PubMed]
  56. Coorssen, J.R.; Padula, M.P. Proteomics—The State of the Field: The Definition and Analysis of Proteomes Should Be Based in Reality, Not Convenience. Proteomes 2024, 12, 14. [Google Scholar] [CrossRef] [PubMed]
  57. Dupree, E.J.; Jayathirtha, M.; Yorkey, H.; Mihasan, M.; Petre, B.A.; Darie, C.C. A Critical Review of Bottom-Up Proteomics: The Good, the Bad, and the Future of this Field. Proteomes 2020, 8, 14. [Google Scholar] [CrossRef]
  58. Xu, T.; Wang, Q.; Wang, Q.; Sun, L. Mass spectrometry-intensive top-down proteomics: An update on technology advancements and biomedical applications. Anal. Methods 2024, 16, 4664–4682. [Google Scholar] [CrossRef] [PubMed]
  59. Carbonara, K.; Andonovski, M.; Coorssen, J.R. Proteomes Are of Proteoforms: Embracing the Complexity. Proteomes 2021, 9, 38. [Google Scholar] [CrossRef] [PubMed]
  60. Hillman, H. Limitations of clinical and biological histology. Med. Hypotheses 2000, 54, 553–564. [Google Scholar] [CrossRef]
  61. Galant, C.; Malghem, J.; Sibille, C.; Docquier, P.-L.; Delloye, C. Current limitations to the histopathological diagnosis of some frequently encountered bone tumours. Acta Orthop. Belg. 2008, 74, 1–6. [Google Scholar]
  62. Soare, I.; Muntean, P.; Mirica, R. Brain Tumors Presumed Malignant without Biopsy-Evaluation and Treatment Problems. Austin J. Clin. Neurol. 2021, 8, 1148. [Google Scholar] [CrossRef]
  63. Zhang, J.; Rector, J.; Lin, J.Q.; Young, J.H.; Sans, M.; Katta, N.; Giese, N.; Yu, W.; Nagi, C.; Suliburk, J.; et al. Nondestructive tissue analysis for ex vivo and in vivo cancer diagnosis using a handheld mass spectrometry system. Sci. Transl. Med. 2017, 9, eaan3968. [Google Scholar] [CrossRef]
  64. Kramer, C.; Vreeswijk, M.; Thijssen, B.; Bosse, T.; Wesseling, J. Beyond the snapshot: Optimizing prognostication and prediction by moving from fixed to functional multidimensional cancer pathology. J. Pathol. 2022, 257, 403–412. [Google Scholar] [CrossRef]
  65. Deng, X.; Yan, Y.; Zhan, Z.; Xie, J.; Tang, H.; Zou, Y.; Tu, J.; Liu, P. A glimpse into the future: Integrating artificial intelligence for precision HER2-positive breast cancer management. iMetaOmics 2024, 1, e19. [Google Scholar] [CrossRef]
  66. Tseng, L.-J.; Matsuyama, A.; MacDonald-Dickinson, V. Histology: The gold standard for diagnosis? Can. Vet. J. 2023, 64, 389–391. [Google Scholar]
  67. Masood, S.; Rosa, M.; Kraemer, D.F.; Smotherman, C.; Mohammadi, A. Comparative cost-effectiveness of fine needle aspiration biopsy versus image-guided biopsy, and open surgical biopsy in the evaluation of breast cancer in the era of affordable care act: A changing landscape. Diagn. Cytopathol. 2015, 43, 605–612. [Google Scholar] [CrossRef]
  68. Silva, E.; Meschter, S.; Tan, M.P. Breast biopsy techniques in a global setting—Clinical practice review. Transl. Breast Cancer Res. 2023, 4, 14. [Google Scholar] [CrossRef]
  69. Wang, M.; He, X.; Chang, Y.; Sun, G.; Thabane, L. A sensitivity and specificity comparison of fine needle aspiration cytology and core needle biopsy in evaluation of suspicious breast lesions: A systematic review and meta-analysis. Breast 2017, 31, 157–166. [Google Scholar] [CrossRef]
  70. Park, H.E.; Han, D.; Lee, J.S.; Nikas, I.P.; Kim, H.; Yang, S.; Lee, H.; Ryu, H.S. Comparison of Breast Fine-Needle Aspiration Cytology and Tissue Sampling for High-Throughput Proteomic Analysis and Cancer Biomarker Detection. Pathobiology 2024, 91, 359–369. [Google Scholar] [CrossRef]
  71. Hwang, W.; Raymond, T.; McPartland, T.; Jeong, S.; Evans, C.L. Fluorescence Lifetime Multiplexing (FLEX) for simultaneous high dimensional spatial biology in 3D. Commun. Biol. 2024, 7, 1012. [Google Scholar] [CrossRef]
  72. Ren, Y.; Wang, M.; Couto, S.; Hansel, D.; Miller, K.; Lopez-Girona, A.; Bjorklund, C.; Gandhi, A.; Thakurta, A.; Chopra, R.; et al. A Dual Color Immunohistochemistry Assay for Measurement of Cereblon in Multiple Myeloma Patient Samples. Appl. Immunohistochem. Mol. Morphol. 2015, 24, 695–702. [Google Scholar] [CrossRef] [PubMed]
  73. Mito, J.K.; Conner, J.R.; Hornick, J.L.; Cibas, E.S.; Qian, X. SOX10/keratin dual-color immunohistochemistry: An effective first-line test for the workup of epithelioid malignant neoplasms in FNA and small biopsy specimens. Cancer Cytopathol. 2018, 126, 179–189. [Google Scholar] [CrossRef] [PubMed]
  74. Rizk, E.M.; Gartrell, R.D.; Barker, L.W.; Esancy, C.L.; Finkel, G.G.; Bordbar, D.D.; Saenger, Y.M. Prognostic and Predictive Immunohistochemistry-Based Biomarkers in Cancer and Immunotherapy. Hematol. Oncol. Clin. N. Am. 2019, 33, 291–299. [Google Scholar] [CrossRef]
  75. Yamashita-Kashima, Y.; Shu, S.; Yorozu, K.; Hashizume, K.; Moriya, Y.; Fujimoto-Ouchi, K.; Harada, N. Importance of formalin fixing conditions for HER2 testing in gastric cancer: Immunohistochemical staining and fluorescence in situ hybridization. Gastric Cancer 2014, 17, 638–647. [Google Scholar] [CrossRef]
  76. Chen, Y.-Y.; Yang, C.-F.; Hsu, C.-Y. The impact of modified staining method on HER2 immunohistochemical staining for HER2-low breast cancer. Pathology 2023, 56, 122–124. [Google Scholar] [CrossRef] [PubMed]
  77. Zafrani, B.; Aubriot, M.-H.; Mouret, E.; De Crémoux, P.; De Rycke, Y.; Nicolas, A.; Boudou, E.; Vincent-Salomon, A.; Magdelénat, H.; Sastre-Garau, X. High sensitivity and specificity of immunohistochemistry for the detection of hormone receptors in breast carcinoma: Comparison with biochemical determination in a prospective study of 793 cases. Histopathology 2000, 37, 536–545. [Google Scholar] [CrossRef]
  78. Thanasan, S.; Sukhakul, K.; Chitpakdee, S.; Kitkumthorn, N. Diagnostic Accuracy of Immunohistochemistry for HER2-Positive Breast Cancer. Asian Pac. J. Cancer Prev. 2023, 24, 4321–4327. [Google Scholar] [CrossRef] [PubMed]
  79. Shamshirian, A.; Aref, A.R.; Yip, G.W.; Warkiani, M.E.; Heydari, K.; Bazaz, S.R.; Hamzehgardeshi, Z.; Shamshirian, D.; Moosazadeh, M.; Alizadeh-Navaei, R. Diagnostic value of serum HER2 levels in breast cancer: A systematic review and meta-analysis. BMC Cancer 2020, 20, 1049. [Google Scholar] [CrossRef] [PubMed]
  80. Gonçalves, J.P.L.; Bollwein, C.; Noske, A.; Jacob, A.; Jank, P.; Loibl, S.; Nekljudova, V.; Fasching, P.A.; Karn, T.; Marmé, F.; et al. Characterization of Hormone Receptor and HER2 Status in Breast Cancer Using Mass Spectrometry Imaging. Int. J. Mol. Sci. 2023, 24, 2860. [Google Scholar] [CrossRef]
  81. Kurreck, A.; Vandergrift, L.A.; Fuss, T.L.; Habbel, P.; Agar, N.Y.R.; Cheng, L.L. Prostate cancer diagnosis and characterization with mass spectrometry imaging. Prostate Cancer Prostatic Dis. 2018, 21, 297–305. [Google Scholar] [CrossRef]
  82. Huang, N.; Chen, L.; He, J.; Nguyen, Q.D. The Efficacy of Clinical Breast Exams and Breast Self-Exams in Detecting Malignancy or Positive Ultrasound Findings. Cureus 2022, 14, e22464. [Google Scholar] [CrossRef]
  83. O’Malley, M.S.; Fletcher, S.W. Screening for Breast Cancer With Breast Self-examination: A Critical Review. JAMA 1987, 257, 2196–2203. [Google Scholar] [CrossRef]
  84. Mango, V.L.; Olasehinde, O.; Omisore, A.D.; Wuraola, F.O.; Famurewa, O.C.; Sevilimedu, V.; Knapp, G.C.; Steinberg, E.; Akinmaye, P.R.; Adewoyin, B.D.; et al. The iBreastExam versus clinical breast examination for breast evaluation in high risk and symptomatic Nigerian women: A prospective study. Lancet Glob. Health 2022, 10, e555–e563. [Google Scholar] [CrossRef]
  85. Bhimani, F.; Zhang, J.; Shah, L.; McEvoy, M.; Gupta, A.; Pastoriza, J.; Shihabi, A.; Feldman, S. Can the Clinical Utility of iBreastExam, a Novel Device, Aid in Optimizing Breast Cancer Diagnosis? A Systematic Review. JCO Glob. Oncol. 2023, 9, e2300149. [Google Scholar] [CrossRef]
  86. Gerami, R.; Joni, S.S.; Akhondi, N.; Etemadi, A.; Fosouli, M.; Eghbal, A.; Souri, Z. A literature review on the imaging methods for breast cancer. Int. J. Physiol. Pathophysiol. Pharmacol. 2022, 14, 171–176. [Google Scholar]
  87. Sood, R.; Rositch, A.F.; Shakoor, D.; Ambinder, E.; Pool, K.-L.; Pollack, E.; Mollura, D.J.; Mullen, L.A.; Harvey, S.C. Ultrasound for Breast Cancer Detection Globally: A Systematic Review and Meta-Analysis. J. Glob. Oncol. 2019, 5, 1–17. [Google Scholar] [CrossRef]
  88. Zeeshan, M.; Salam, B.; Khalid, Q.S.; Alam, S.; Sayani, R. Diagnostic Accuracy of Digital Mammography in the Detection of Breast Cancer. Cureus 2018, 10, e2448. [Google Scholar] [CrossRef]
  89. Merriel, S.W.; Akter, N.; Zakkak, N.; Swann, R.; McPhail, S.; Rubin, G.; Lyratzopoulos, G.; Abel, G. Factors affecting prostate cancer detection through asymptomatic prostate-specific antigen testing in primary care in England: Evidence from the 2018 National Cancer Diagnosis Audit. Br. J. Gen. Pract. 2024, 75, e300–e305. [Google Scholar] [CrossRef]
  90. Uematsu, T. Sensitivity and specificity of screening mammography without clinical breast examination among Japanese women aged 40–49 years: Analysis of data from the J-START results. Breast Cancer 2022, 29, 928–931. [Google Scholar] [CrossRef] [PubMed]
  91. Gøtzsche, P.C. Mammography screening is harmful and should be abandoned. J. R. Soc. Med. 2015, 108, 341–345. [Google Scholar] [CrossRef] [PubMed]
  92. Garg, M.; Karami, V.; Moazen, J.; Kwee, T.; Bhalla, A.S.; Shahbazi-Gahrouei, D.; Shao, Y.-H.J. Radiation Exposure and Lifetime Attributable Risk of Cancer Incidence and Mortality from Low- and Standard-Dose CT Chest: Implications for COVID-19 Pneumonia Subjects. Diagnostics 2022, 12, 3043. [Google Scholar] [CrossRef]
  93. Cao, C.-F.; Ma, K.-L.; Shan, H.; Liu, T.-F.; Zhao, S.-Q.; Wan, Y.; Jun, Z.; Wang, H.-Q. CT Scans and Cancer Risks: A Systematic Review and Dose-response Meta-analysis. BMC Cancer 2022, 22, 1238. [Google Scholar] [CrossRef] [PubMed]
  94. Schultz, C.H.; Fairley, R.; Murphy, L.S.-L.; Doss, M. The Risk of Cancer from CT Scans and Other Sources of Low-Dose Radiation: A Critical Appraisal of Methodologic Quality. Prehospital Disaster Med. 2020, 35, 3–16. [Google Scholar] [CrossRef]
  95. Younis, S.; Emayof, M. Role CT imaging for Cancer Diagnosis by Determining Sensitivity and Specificity: 144 Cancer Cases in Tobruk, Libya. AlQalam J. Med. Appl. Sci. 2024, 7, 1458–1463. [Google Scholar] [CrossRef]
  96. Toyoda, Y.; Nakayama, T.; Kusunoki, Y.; Iso, H.; Suzuki, T. Sensitivity and specificity of lung cancer screening using chest low-dose computed tomography. Br. J. Cancer 2008, 98, 1602–1607. [Google Scholar] [CrossRef]
  97. Cochran, T.; Patel, S.; Kruse, T.; Lyden, E.; Comer, S.; Ford, J. Sensitivity and Specificity of Whole-body MRI for the Detection of Pediatric Malignancy. J. Pediatr. Hematol. Oncol. 2023, 45, e26–e30. [Google Scholar] [CrossRef]
  98. Aziz, S.; Mohamad, M.; Zin, R. Histopathological Correlation of Breast Carcinoma with Breast Imaging-Reporting and Data System. Malays. J. Med. Sci. 2022, 29, 65–74. [Google Scholar] [CrossRef]
  99. Niikura, N.; Costelloe, C.M.; Madewell, J.E.; Hayashi, N.; Yu, T.K.; Liu, J.; Palla, S.L.; Tokuda, Y.; Theriault, R.L.; Hortobagyi, G.N.; et al. FDG-PET/CT Compared with Conventional Imaging in the Detection of Distant Metastases of Primary Breast Cancer. Oncologist 2011, 16, 1111–1119. [Google Scholar] [CrossRef]
  100. Butt, F.; Dominguez-Konicki, L.; Tocci, N.; Paydarfar, J.; Seltzer, M.; Pastel, D. Diagnostic accuracy of the latest-generation digital PET/CT scanner for detection of metastatic lymph nodes in head and neck cancer. Front. Nucl. Med. 2023, 3, 1184448. [Google Scholar] [CrossRef] [PubMed]
  101. Vetrone, L.; Mei, R.; Bianchi, L.; Giunchi, F.; Farolfi, A.; Castellucci, P.; Droghetti, M.; Presutti, M.; Degiovanni, A.; Schiavina, R.; et al. Histology and PSMA Expression on Immunohistochemistry in High-Risk Prostate Cancer Patients: Comparison with 68Ga-PSMA PET/CT Features in Primary Staging. Cancers 2023, 15, 1716. [Google Scholar] [CrossRef]
  102. Houshmand, S.; Lawhn-Heath, C.; Behr, S. PSMA PET imaging in the diagnosis and management of prostate cancer. Abdom. Radiol. 2023, 48, 3610–3623. [Google Scholar] [CrossRef]
  103. von Stauffenberg, F.; Poyet, C.; Beintner-Skawran, S.; Maurer, A.; Schmid, F.A. Current Clinical Applications of PSMA-PET for Prostate Cancer Diagnosis, Staging, and Treatment. Cancers 2024, 16, 4263. [Google Scholar] [CrossRef] [PubMed]
  104. Belal, S.L.; Frantz, S.; Minarik, D.; Enqvist, O.; Wikström, E.; Edenbrandt, L.; Trägårdh, E. Applications of Artificial Intelligence in PSMA PET/CT for Prostate Cancer Imaging. Semin. Nucl. Med. 2024, 54, 141–149. [Google Scholar] [CrossRef] [PubMed]
  105. Ionescu, F.; Zhang, J.; Wang, L. Clinical Applications of Liquid Biopsy in Prostate Cancer: From Screening to Predictive Biomarker. Cancers 2022, 14, 1728. [Google Scholar] [CrossRef]
  106. Frantzi, M.; Culig, Z.; Heidegger, I.; Mokou, M.; Latosinska, A.; Roesch, M.C.; Merseburger, A.S.; Makridakis, M.; Vlahou, A.; Blanca-Pedregosa, A.; et al. Mass Spectrometry-Based Biomarkers to Detect Prostate Cancer: A Multicentric Study Based on Non-Invasive Urine Collection without Prior Digital Rectal Examination. Cancers 2023, 15, 1166. [Google Scholar] [CrossRef]
  107. Höti, N.; Lih, T.-S.; Dong, M.; Zhang, Z.; Mangold, L.; Partin, A.W.; Sokoll, L.J.; Li, Q.K.; Zhang, H. Urinary PSA and Serum PSA for Aggressive Prostate Cancer Detection. Cancers 2023, 15, 960. [Google Scholar] [CrossRef]
  108. Tanase, C.P.; Codrici, E.; Popescu, I.D.; Mihai, S.; Enciu, A.-M.; Necula, L.G.; Preda, A.; Ismail, G.; Albulescu, R. Prostate cancer proteomics: Current trends and future perspectives for biomarker discovery. Oncotarget 2017, 8, 18497. [Google Scholar] [CrossRef] [PubMed]
  109. Luo, Z.-H.; Lu, P.-X.; Qi, W.-L.; Jin, A.-F.; Liu, Q.; Zeng, Q.-Y.; Lu, P. The sensitivity and specificity of 18F-FDG PET/CT in spinal leptomeningeal metastases: The synergistic effect of the 18F-FDG PET-CT to gadolinium-enhanced MRI. Quant. Imaging Med. Surg. 2023, 13, 6863–6875. [Google Scholar] [CrossRef] [PubMed]
  110. Bashir, U.; Mallia, A.; Stirling, J.; Joemon, J.; MacKewn, J.; Charles-Edwards, G.; Goh, V.; Cook, G.J. PET/MRI in Oncological Imaging: State of the Art. Diagnostics 2015, 5, 333–357. [Google Scholar] [CrossRef]
  111. Kitajima, K.; Nakamoto, Y.; Okizuka, H.; Onishi, Y.; Senda, M.; Suganuma, N.; Sugimura, K. Accuracy of whole-body FDG-PET/CT for detecting brain metastases from non-central nervous system tumors. Ann. Nucl. Med. 2008, 22, 595–602. [Google Scholar] [CrossRef] [PubMed]
  112. Yang, T.; Li, W.; Huang, T.; Zhou, J. Genetic Testing Enhances the Precision Diagnosis and Treatment of Breast Cancer. Int. J. Mol. Sci. 2023, 24, 16607. [Google Scholar] [CrossRef]
  113. Lynce, F.; Isaacs, C. How Far Do We Go With Genetic Evaluation? Gene, Panel, and Tumor Testing. Am. Soc. Clin. Oncol. Educ. Book 2016, 35, e72–e78. [Google Scholar] [CrossRef]
  114. Venetis, K.; Pescia, C.; Cursano, G.; Frascarelli, C.; Mane, E.; De Camilli, E.; Munzone, E.; Dellapasqua, S.; Criscitiello, C.; Curigliano, G.; et al. The Evolving Role of Genomic Testing in Early Breast Cancer: Implications for Diagnosis, Prognosis, and Therapy. Int. J. Mol. Sci. 2024, 25, 5717. [Google Scholar] [CrossRef]
  115. Yamamoto, S.; Chishima, T.; Shibata, Y.; Harada, F.; Takeuchi, H.; Yamada, A.; Narui, K.; Misumi, T.; Ishikawa, T.; Endo, I. Clinical Impact of a Novel Model Predictive of Oncotype DX Recurrence Score in Breast Cancer. In Vivo 2021, 35, 2439. [Google Scholar] [CrossRef] [PubMed]
  116. Lee, Y.J.; Hwang, Y.S.; Kim, J.; Ahn, S.-H.; Son, B.H.; Kim, H.J.; Ko, B.S.; Kim, J.; Chung, I.Y.; Lee, J.W.; et al. A nomogram for predicting probability of low risk of MammaPrint results in women with clinically high-risk breast cancer. Sci. Rep. 2021, 11, 23509. [Google Scholar] [CrossRef]
  117. Wallden, B.; Storhoff, J.; Nielsen, T.; Dowidar, N.; Schaper, C.; Ferree, S.; Liu, S.; Leung, S.; Geiss, G.; Snider, J.; et al. Development and verification of the PAM50-based Prosigna breast cancer gene signature assay. BMC Med. Genom. 2015, 8, 54. [Google Scholar] [CrossRef]
  118. Dubsky, P.C.; Singer, C.F.; Egle, D.; Wette, V.; Petru, E.; Balic, M.; Pichler, A.; Greil, R.; Petzer, A.L.; Bago-Horvath, Z.; et al. The EndoPredict score predicts response to neoadjuvant chemotherapy and neoendocrine therapy in hormone receptor-positive, human epidermal growth factor receptor 2-negative breast cancer patients from the ABCSG-34 trial. Eur. J. Cancer 2020, 134, 99–106. [Google Scholar] [CrossRef]
  119. Bartlett, J.M.S.; Sgroi, D.C.; Treuner, K.; Zhang, Y.; Piper, T.; Salunga, R.C.; Ahmed, I.; Doos, L.; Thornber, S.; Taylor, K.J.; et al. Breast Cancer Index Is a Predictive Biomarker of Treatment Benefit and Outcome from Extended Tamoxifen Therapy: Final Analysis of the Trans-aTTom Study. Clin. Cancer Res. 2022, 28, 1871–1880. [Google Scholar] [CrossRef]
  120. Leapman, M.S.; Sutherland, R.; Gross, C.P.; Ma, X.; Seibert, T.M.; Cooperberg, M.R.; Catalona, W.J.; Loeb, S.; Schulman-Green, D. Patient experiences with tissue-based genomic testing during active surveillance for prostate cancer. BJUI Compass 2024, 5, 142–149. [Google Scholar] [CrossRef]
  121. McHugh, J.K.; Bancroft, E.K.; Saunders, E.; Brook, M.N.; McGrowder, E.; Wakerell, S.; James, D.; Rageevakumar, R.; Benton, B.; Taylor, N.; et al. Assessment of a Polygenic Risk Score in Screening for Prostate Cancer. N. Engl. J. Med. 2025, 392, 1406–1417. [Google Scholar] [CrossRef]
  122. Katsuya, Y. Current and future trends in whole genome sequencing in cancer. Cancer Biol. Med. 2024, 21, 16. [Google Scholar] [CrossRef]
  123. Thierry, A.R. Circulating DNA fragmentomics and cancer screening. Cell Genom. 2023, 3, 100242. [Google Scholar] [CrossRef] [PubMed]
  124. Pessoa, L.S.; Heringer, M.; Ferrer, V.P. ctDNA as a cancer biomarker: A broad overview. Crit. Rev. Oncol. Hematol. 2020, 155, 103109. [Google Scholar] [CrossRef] [PubMed]
  125. Pritchard, D.; Goodman, C.; Nadauld, L.D. Clinical Utility of Genomic Testing in Cancer Care. JCO Precis. Oncol. 2022, 6, e2100349. [Google Scholar] [CrossRef]
  126. Akhoundova, D.; Rubin, M. The grand challenge of moving cancer whole-genome sequencing into the clinic. Nat. Med. 2024, 30, 39–40. [Google Scholar] [CrossRef]
  127. Rebello, R.; Posner, A.; Dong, R.; Prall, O.; Sivakumaran, T.; Mitchell, C.; Flynn, A.; Caneborg, A.; Mitchell, C.; Kanwal, S.; et al. Whole genome sequencing improves tissue-of-origin diagnosis and treatment options for cancer of unknown primary. Nat. Commun. 2025, 16, 4422. [Google Scholar] [CrossRef] [PubMed]
  128. Zalis, M.; Veloso, G.G.V.; Aguiar, P.N., Jr.; Gimenes, N.; Reis, M.X.; Matsas, S.; Ferreira, C.G. Next-generation sequencing impact on cancer care: Applications, challenges, and future directions. Front. Genet. 2024, 15, 1420190. [Google Scholar] [CrossRef]
  129. Donohue, K.E.; Gooch, C.; Katz, A.; Wakelee, J.; Slavotinek, A.; Korf, B.R. Pitfalls and challenges in genetic test interpretation: An exploration of genetic professionals experience with interpretation of results. Clin. Genet. 2021, 99, 638–649. [Google Scholar] [CrossRef]
  130. Casolino, R.; Beer, P.A.; Chakravarty, D.; Davis, M.B.; Malapelle, U.; Mazzarella, L.; Normanno, N.; Pauli, C.; Subbiah, V.; Turnbull, C.; et al. Interpreting and integrating genomic tests results in clinical cancer care: Overview and practical guidance. CA Cancer J. Clin. 2024, 74, 264–285. [Google Scholar] [CrossRef]
  131. Zhou, Y.; Tao, L.; Qiu, J.; Xu, J.; Yang, X.; Zhang, Y.; Tian, X.; Guan, X.; Cen, X.; Zhao, Y. Tumor biomarkers for diagnosis, prognosis and targeted therapy. Signal Transduct. Target. Ther. 2024, 9, 132. [Google Scholar] [CrossRef] [PubMed]
  132. Ahmed, M.; Wuethrich, A.; Constantin, N.; Shanmugasundaram, K.B.; Mainwaring, P.; Kulasinghe, A.; O’Leary, C.; O’Byrne, K.; Sina, A.A.I.; Carrascosa, L.G.; et al. Liquid Biopsy Snapshots of Key Phosphoproteomic Pathways in Lung Cancer Patients for Diagnosis and Therapy Monitoring. Anal. Chem. 2023, 95, 8522–8532. [Google Scholar] [CrossRef]
  133. Neagu, A.-N.; Jayathirtha, M.; Whitham, D.; Mutsengi, P.; Sullivan, I.; Petre, B.A.; Darie, C.C. Proteomics-Based Identification of Dysregulated Proteins in Breast Cancer. Proteomes 2022, 10, 35. [Google Scholar] [CrossRef] [PubMed]
  134. Agrawal, K.; Shi, Y.; Del Rosario, A.M.; Jian, W. Method Development Workflow for Quantifying Protein Biomarkers by Hybrid LC–MS/MS. Bioanalysis 2022, 14, 985–1004. [Google Scholar] [CrossRef]
  135. Macklin, A.; Khan, S.; Kislinger, T. Recent advances in mass spectrometry based clinical proteomics: Applications to cancer research. Clin. Proteom. 2020, 17, 17. [Google Scholar] [CrossRef]
  136. Jiang, W.; Wei, W.; Krishnamoorthy, G.; Hu, P.; Chen, M.; Tiedje, V.; Acuña-Ruiz, A.; Wang, H.; Wang, Z.; Wang, J.; et al. Comprehensive Mass Spectral Libraries of Human Thyroid Tissues and Cells. Sci. Data 2024, 11, 1448. [Google Scholar] [CrossRef]
  137. Montero, A.; Garranzo-Asensio, M.; Rejas-González, R.; Feliu, J.; Mendiola, M.; García, A.; Barderas, R. Benefits of FAIMS to Improve the Proteome Coverage of Deteriorated and/or Cross-Linked TMT 10-Plex FFPE Tissue and Plasma-Derived Exosomes Samples. Proteomes 2023, 11, 35. [Google Scholar] [CrossRef]
  138. Lu, P.; Yang, L.; Chen, W.; Li, K.; Chen, X.; Qu, S. Four-dimensional trapped ion mobility spectrometry proteomics reveals circulating extracellular vesicles encapsulated drivers of nasopharyngeal carcinoma distant dissemination. Talanta 2025, 282, 126907. [Google Scholar] [CrossRef] [PubMed]
  139. Brzhozovskiy, A.; Kononikhin, A.; Bugrova, A.E.; Kovalev, G.I.; Schmit, P.-O.; Kruppa, G.; Nikolaev, E.N.; Borchers, C.H. The Parallel Reaction Monitoring-Parallel Accumulation–Serial Fragmentation (prm-PASEF) Approach for Multiplexed Absolute Quantitation of Proteins in Human Plasma. Anal. Chem. 2022, 94, 2016–2022. [Google Scholar] [CrossRef]
  140. Bradley, S.; Watson, J. Testing and cancer diagnosis in general practice. BMJ Qual. Amp. Saf. 2025, 34, 351–354. [Google Scholar] [CrossRef] [PubMed]
  141. Naji, L.; Randhawa, H.; Sohani, Z.; Dennis, B.; Lautenbach, D.; Kavanagh, O.; Bawor, M.; Banfield, L.; Profetto, J. Digital Rectal Examination for Prostate Cancer Screening in Primary Care: A Systematic Review and Meta-Analysis. Ann. Fam. Med. 2018, 16, 149–154. [Google Scholar] [CrossRef]
  142. Streicher, J.; Meyerson, B.L.; Karivedu, V.; Sidana, A. A review of optimal prostate biopsy: Indications and techniques. Ther. Adv. Urol. 2019, 11, 1756287219870074. [Google Scholar] [CrossRef]
  143. Reza, H.S.; Ali, Z.; Tara, H.; Ali, B. Age-specific reference ranges of prostate-specific antigen in the elderly of Amirkola: A population-based study. Asian J. Urol. 2021, 8, 183–188. [Google Scholar] [CrossRef]
  144. Chen, R.; Huang, Y.; Cai, X.; Xie, L.; He, D.; Zhou, L.; Xu, C.; Gao, X.; Ren, S.; Wang, F.; et al. Age-Specific Cutoff Value for the Application of Percent Free Prostate-Specific Antigen (PSA) in Chinese Men with Serum PSA Levels of 4.0–10.0 ng/mL. PLoS ONE 2015, 10, e0130308. [Google Scholar] [CrossRef]
  145. Aref, A.T.; Vincent, A.D.; O’Callaghan, M.E.; Martin, S.A.; Sutherland, P.D.; Hoy, A.J.; Butler, L.M.; Wittert, G.A. The inverse relationship between prostate specific antigen (PSA) and obesity. Endocr. Relat. Cancer 2018, 25, 933–941. [Google Scholar] [CrossRef] [PubMed]
  146. Koc, G.; Akgul, K.; Yilmaz, Y.; Dirik, A. The effects of cigarette smoking on prostate-specific antigen in two different age groups. Can. Urol. Assoc. J. 2013, 7, e704–e707. [Google Scholar] [CrossRef] [PubMed]
  147. Press, D.J.; Pierce, B.; Lauderdale, D.S.; Aschebrook-Kilfoy, B.; Gomez, S.L.; Hedeker, D.; Wright, N.E.; Fantus, R.J.; Bettencourt, L.; Ahsan, H.; et al. Tobacco and marijuana use and their association with serum prostate-specific antigen levels among African American men in Chicago. Prev. Med. Rep. 2020, 20, 101174. [Google Scholar] [CrossRef]
  148. Barlow, M.; Down, L.; Mounce, L.T.A.; Merriel, S.W.D.; Watson, J.; Martins, T.; Bailey, S.E.R. Ethnic differences in prostate-specific antigen levels in men without prostate cancer: A systematic review. Prostate Cancer Prostatic Dis. 2023, 26, 249–256. [Google Scholar] [CrossRef]
  149. Down, L.; Barlow, M.; Bailey, S.E.R.; Mounce, L.T.A.; Merriel, S.W.D.; Watson, J.; Martins, T. Association between patient ethnicity and prostate cancer diagnosis following a prostate-specific antigen test: A cohort study of 730,000 men in primary care in the UK. BMC Med. 2024, 22, 82. [Google Scholar] [CrossRef]
  150. Urabe, F.; Sumiyoshi, T.; Tashiro, K.; Goto, T.; Kimura, T.; Kobayashi, T. Prostate cancer and liquid biopsies: Clinical applications and challenges. Int. J. Urol. 2024, 31, 617–626. [Google Scholar] [CrossRef]
  151. Joshi, N.; Garapati, K.; Ghose, V.; Kandasamy, R.K.; Pandey, A. Recent progress in mass spectrometry-based urinary proteomics. Clin. Proteom. 2024, 21, 14. [Google Scholar] [CrossRef] [PubMed]
  152. Wang, G.; Jin, W.; Xu, Z.; Ju, L.; Shan, D.; Li, S.; Yu, M.; Cao, X.; Liu, N.; Qian, K.; et al. Urine-based liquid biopsy in bladder cancer: Opportunities and challenges. Clin. Transl. Discov. 2023, 3, e176. [Google Scholar] [CrossRef]
  153. Zhou, X.; Xue, F.; Li, T.; Xue, J.; Yue, S.; Zhao, S.; Lu, H.; He, C. Exploration of potential biomarkers for early bladder cancer based on urine proteomics. Front. Oncol. 2024, 14, 1309842. [Google Scholar] [CrossRef]
  154. Ha, A.; Khoo, A.; Ignatchenko, V.; Khan, S.; Waas, M.; Vesprini, D.; Liu, S.K.; Nyalwidhe, J.O.; Semmes, O.J.; Boutros, P.C.; et al. Comprehensive Prostate Fluid-Based Spectral Libraries for Enhanced Protein Detection in Urine. J. Proteome Res. 2024, 23, 1768–1778. [Google Scholar] [CrossRef] [PubMed]
  155. Lee, J.; McKinney, K.; Pavlopoulos, A.; Niu, M.; Kang, J.; Oh, J.; Kim, K.P.; Hwang, S. Altered Proteome of Extracellular Vesicles Derived from Bladder Cancer Patients Urine. Mol. Cells 2018, 41, 179–187. [Google Scholar] [CrossRef]
  156. Beasley-Green, A. Urine Proteomics in the Era of Mass Spectrometry. Int. Neurourol. J. 2016, 20, S70–S75. [Google Scholar] [CrossRef]
  157. Wood, S.L.; Knowles, M.A.; Thompson, D.; Selby, P.J.; Banks, R.E. Proteomic studies of urinary biomarkers for prostate, bladder and kidney cancers. Nat. Rev. Urol. 2013, 10, 206–218. [Google Scholar] [CrossRef]
  158. Jordaens, S.; Zwaenepoel, K.; Tjalma, W.; Deben, C.; Beyers, K.; Vankerckhoven, V.; Pauwels, P.; Vorsters, A. Urine biomarkers in cancer detection: A systematic review of preanalytical parameters and applied methods. Int. J. Cancer 2023, 152, 2186–2205. [Google Scholar] [CrossRef] [PubMed]
  159. Albakova, Z.; Norinho, D.D.; Mangasarova, Y.; Sapozhnikov, A. Heat Shock Proteins in Urine as Cancer Biomarkers. Front. Med. 2021, 8, 743476. [Google Scholar] [CrossRef]
  160. Ramirez-Garrastacho, M.; Bajo-Santos, C.; Line, A.; Martens-Uzunova, E.S.; de la Fuente, J.M.; Moros, M.; Soekmadji, C.; Tasken, K.A.; Llorente, A. Extracellular vesicles as a source of prostate cancer biomarkers in liquid biopsies: A decade of research. Br. J. Cancer 2022, 126, 331–350. [Google Scholar] [CrossRef]
  161. Hamid, Y.; Rabbani, R.D.; Afsara, R.; Nowrin, S.; Ghose, A.; Papadopoulos, V.; Sirlantzis, K.; Ovsepian, S.V.; Boussios, S. Exosomal Liquid Biopsy in Prostate Cancer: A Systematic Review of Biomarkers for Diagnosis, Prognosis, and Treatment Response. Int. J. Mol. Sci. 2025, 26, 802. [Google Scholar] [CrossRef]
  162. Li, S.; Yi, M.; Dong, B.; Tan, X.; Luo, S.; Wu, K. The role of exosomes in liquid biopsy for cancer diagnosis and prognosis prediction. Int. J. Cancer 2021, 148, 2640–2651. [Google Scholar] [CrossRef] [PubMed]
  163. Cristina Barbosa da Silva, A.C.B.; Albuquerque, A.; Romario-Silva, D.; Ferreira, S.; Rodrigues, A.; Marinho, S. Salivary Diagnostics, Current Reality and Future Prospects. In Emerging Trends in Oral Health Sciences and Dentistry; Virdi, M.S., Ed.; IntechOpen: Rijeka, Croatia, 2015. [Google Scholar] [CrossRef]
  164. Limeres, J.; Dios, P.; Scully, C. Infection Transmission by Saliva and the Paradoxical Protective Role of Saliva. Saliva Prot. Transm. Dis. 2017, 1, 1–18. [Google Scholar] [CrossRef]
  165. Dame, Z.T.; Aziat, F.; Mandal, R.; Krishnamurthy, R.; Bouatra, S.; Borzouie, S.; Guo, A.C.; Sajed, T.; Deng, L.; Lin, H.; et al. The human saliva metabolome. Metabolomics 2015, 11, 1864–1883. [Google Scholar] [CrossRef]
  166. Subbarao, K.C.; Nattuthurai, G.S.; Sundararajan, S.K.; Sujith, I.; Joseph, J.; Syedshah, Y.P. Gingival Crevicular Fluid: An Overview. J. Pharm. Bioallied Sci. 2019, 11, S135–S139. [Google Scholar] [CrossRef]
  167. Das, A.; Bhattacharjee, S. Saliva as a sample for liquid biopsy: A review. J. Oral Med. Oral Surg. Oral Pathol. Oral Radiol. 2024, 10, 261–264. [Google Scholar] [CrossRef]
  168. Koopaie, M.; Kolahdooz, S.; Fatahzadeh, M.; Manifar, S. Salivary biomarkers in breast cancer diagnosis: A systematic review and diagnostic meta-analysis. Cancer Med. 2022, 11, 2644–2661. [Google Scholar] [CrossRef]
  169. Rashid, S.; Puttagunta, P.; Pamulapati, S.; Yang, J.; Pocha, S.; Saba, N.F.; Teng, Y. Leveraging Saliva for Insights into Head and Neck Cancer. Int. J. Mol. Sci. 2024, 25, 13514. [Google Scholar] [CrossRef] [PubMed]
  170. Krapfenbauer, K.; Drucker, E.; Thurnher, D. Identification of tumour-related proteins as potential screening markers by proteome analysis—Protein profiles of human saliva as a predictive and prognostic tool. EPMA J. 2014, 5, 20. [Google Scholar] [CrossRef]
  171. Archana, N.; Atharva, D. Salivary Biomarkers for Oral Cancer Detection: Insights from Human DNA and RNA Analysis. Cardiovasc. Hematol. Agents Med. Chem. 2024, 22, 249–257. [Google Scholar] [CrossRef]
  172. Suragimath, G.; Patil, S.; Suragimath, D.G.; Sr, A. Salivaomics: A Revolutionary Non-invasive Approach for Oral Cancer Detection. Cureus 2024, 16, e74381. [Google Scholar] [CrossRef]
  173. Gupta, S.; Brijesh, S.; Rajul, A.; Sameer, G.; Sachan, M. The emerging role of liquid biopsy in oral squamous cell carcinoma detection: Advantages and challenges. Expert Rev. Mol. Diagn. 2024, 24, 311–331. [Google Scholar] [CrossRef] [PubMed]
  174. Naito, Y.; Honda, K. Liquid Biopsy for Oral Cancer Diagnosis: Recent Advances and Challenges. J. Pers. Med. 2023, 13, 303. [Google Scholar] [CrossRef] [PubMed]
  175. Bastías, D.; Maturana, A.; Marín, C.; Martínez, R.; Niklander, S.E. Salivary Biomarkers for Oral Cancer Detection: An Exploratory Systematic Review. Int. J. Mol. Sci. 2024, 25, 2634. [Google Scholar] [CrossRef]
  176. Ghallab, N.A.; Shaker, O.G. Serum and salivary levels of chemerin and MMP-9 in oral squamous cell carcinoma and oral premalignant lesions. Clin. Oral Investig. 2017, 21, 937–947. [Google Scholar] [CrossRef] [PubMed]
  177. Aslebagh, R.; Channaveerappa, D.; Arcaro, K.F.; Darie, C.C. Proteomics analysis of human breast milk to assess breast cancer risk. Electrophoresis 2018, 39, 653–665. [Google Scholar] [CrossRef]
  178. Aslebagh, R.; Whitham, D.; Channaveerappa, D.; Mutsengi, P.; Pentecost, B.T.; Arcaro, K.F.; Darie, C.C. Mass Spectrometry-Based Proteomics of Human Milk to Identify Differentially Expressed Proteins in Women with Breast Cancer versus Controls. Proteomes 2022, 10, 36. [Google Scholar] [CrossRef]
  179. Aslebagh, R.; Whitham, D.; Channaveerappa, D.; Lowe, J.; Pentecost, B.T.; Arcaro, K.F.; Darie, C.C. Proteomics analysis of human breast milk by two-dimensional polyacrylamide gel electrophoresis (2D-PAGE) coupled with mass spectrometry to assess breast cancer risk. Electrophoresis 2023, 44, 1097–1113. [Google Scholar] [CrossRef]
  180. Aslebagh, R.; Channaveerappa, D.; Arcaro, K.F.; Darie, C.C. Comparative two-dimensional polyacrylamide gel electrophoresis (2D-PAGE) of human milk to identify dysregulated proteins in breast cancer. Electrophoresis 2018, 39, 1723–1734. [Google Scholar] [CrossRef]
  181. Whitham, D.; Aslebagh, R.; Channaveerappa, D.; Pentecost, B.; Arcaro, K.F.; Darie, C.C. Proteomic Analysis of Human Breast Milk to Reveal Potential Protein Biomarkers for Breast Cancer. FASEB J. 2022, 36. [Google Scholar] [CrossRef]
  182. Durán-Jara, E.; Vera-Tobar, T.; Lobos-González, L.D.L. Lactadherin: From a Well-Known Breast Tumor Marker to a Possible Player in Extracellular Vesicle-Mediated Cancer Progression. Int. J. Mol. Sci. 2022, 23, 3855. [Google Scholar] [CrossRef] [PubMed]
  183. Karav, S. Chapter 22-Application of a Novel Endo-β-N-Acetylglucosaminidase to Isolate an Entirely New Class of Bioactive Compounds: N-Glycans. In Enzymes in Food Biotechnology; Kuddus, M., Ed.; Academic Press: Cambridge, UK, 2019; pp. 389–404. [Google Scholar] [CrossRef]
  184. Gopal, S.H.; Das, S. Role of Lactoferrin in the Carcinogenesis of Triple-Negative Breast Cancer. J. Cancer Clin. Trials 2016, 1, e105. [Google Scholar] [PubMed]
  185. Aslebagh, R.; Channaveerappa, D.; Pentecost, B.T.; Arcaro, K.F.; Darie, C.C. Combinatorial Electrophoresis and Mass Spectrometry-Based Proteomics in Breast Milk for Breast Cancer Biomarker Discovery. In Advancements of Mass Spectrometry in Biomedical Research; Woods, A.G., Darie, C.C., Eds.; Springer International Publishing: Cham, Switzerland, 2019; pp. 451–467. [Google Scholar] [CrossRef]
  186. Keskin, M.; Kompuinen, J.; Harmankaya, İ.; Karaçetin, D.; Nissilä, V.; Gürsoy, M.; Sorsa, T.; Gürsoy, U.K. Oral Cavity Calprotectin and Lactoferrin Levels in Relation to Radiotherapy. Curr. Issues Mol. Biol. 2022, 44, 4439–4446. [Google Scholar] [CrossRef]
  187. Wang, Y.; Bi, X.; Luo, Z.; Wang, H.; Ismtula, D.; Guo, C. Gelsolin: A comprehensive pan-cancer analysis of potential prognosis, diagnostic, and immune biomarkers. Front. Genet. 2023, 14, 1093163. [Google Scholar] [CrossRef]
  188. Akkinepally, H.; Tejasvi, M.A.; Kavitha, N.L.; Pokala, A.; Reddy, P.; Keerthi, V.R. Estimation of Alpha-amylase in Smokers with and without Leukoplakia and Oral Cancer—A Comparative Study. J. Indian Acad. Oral Med. Radiol. 2023, 35, 331–334. [Google Scholar] [CrossRef]
  189. Pierzynowska, K.; Thomasson, S.; Oredsson, S. Alpha-Amylase Inhibits Cell Proliferation and Glucose Uptake in Human Neuroblastoma Cell Lines. BioMed Res. Int. 2022, 2022, 4271358. [Google Scholar] [CrossRef]
  190. Burgos-Panadero, R.; Noguera, I.; Cañete, A.; Navarro, S.; Noguera, R. Vitronectin as a molecular player of the tumor microenvironment in neuroblastoma. BMC Cancer 2019, 19, 479. [Google Scholar] [CrossRef]
  191. Jiwa, N.; Ezzat, A.; Holt, J.; Wijayatilake, D.S.; Takats, Z.; Leff, D.R. Nipple aspirate fluid and its use for the early detection of breast cancer. Ann. Med. Surg. 2022, 77, 103625. [Google Scholar] [CrossRef]
  192. Paweletz, C.; Trock, B.; Pennanen, M.; Tsangaris, T.; Magnant, C.; Liotta, L.; Petricoin, E. Proteomic Patterns of Nipple Aspirate Fluids Obtained by SELDI-TOF: Potential for New Biomarkers to Aid in the Diagnosis of Breast Cancer. Dis. Markers 2001, 17, 301–307. [Google Scholar] [CrossRef] [PubMed]
  193. 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 2021, 14, 159. [Google Scholar] [CrossRef] [PubMed]
  194. Sorolla, M.A.; Sorolla, A.; Parisi, E.; Salud, A.; Porcel, J.M. Diving into the Pleural Fluid: Liquid Biopsy for Metastatic Malignant Pleural Effusions. Cancers 2021, 13, 2798. [Google Scholar] [CrossRef]
  195. Botana-Rial, M.; Vázquez-Iglesias, L.; Casado-Rey, P.; Cadena, M.P.d.l.; Andrade-Olivié, M.A.; Abal-Arca, J.; García-Nimo, L.; Ferreiro-Fernández, L.; Valdés-Cuadrado, L.; San-José, M.E.; et al. Validation of Calprotectin As a Novel Biomarker For The Diagnosis of Pleural Effusion: A Multicentre Trial. Sci. Rep. 2020, 10, 5679. [Google Scholar] [CrossRef]
  196. Nandi, S.K.; Singh, D.; Upadhay, J.; Gupta, N.; Dhiman, N.; Mittal, S.K.; Mahindroo, N. Identification of tear-based protein and non-protein biomarkers: Its application in diagnosis of human diseases using biosensors. Int. J. Biol. Macromol. 2021, 193, 838–846. [Google Scholar] [CrossRef] [PubMed]
  197. Daily, A.; Ravishankar, P.; Harms, S.; Klimberg, V. Using tears as a non-invasive source for early detection of breast cancer. PLoS ONE 2022, 17, e0267676. [Google Scholar] [CrossRef]
  198. Ravishankar, P.; Daily, A. Tears as the Next Diagnostic Biofluid: A Comparative Study between Ocular Fluid and Blood. Appl. Sci. 2022, 12, 2884. [Google Scholar] [CrossRef]
  199. Daily, A.; Ravishankar, P.; Wang, W.; Krone, R.; Harms, S.; Klimberg, V.S. Development and validation of a short-term breast health measure as a supplement to screening mammography. Biomark. Res. 2022, 10, 76. [Google Scholar] [CrossRef]
  200. Kaufmann, Y.; Byrum, S.D.; Acott, A.A.; Siegel, E.R.; Washam, C.L.; Klimberg, V.S.; Mancino, A.T. Proteomic profiling of tear fluid as a promising non-invasive screening test for colon cancer. Am. J. Surg. 2022, 224, 19–24. [Google Scholar] [CrossRef]
  201. Schulz, B.; Oxley, D.; Packer, N.; Karlsson, N. Identification of two highly sialylated human tear-fluid DMBT1 isoforms: The major high-molecular-mass glycoproteins in human tears. Biochem. J. 2002, 366, 511–520. [Google Scholar] [CrossRef]
  202. Midena, E.; Parrozzani, R.; Midena, G.; Trainiti, S.; Marchione, G.; Cosmo, E.; Londei, D.; Frizziero, L. In vivo intraocular biomarkers: Changes of aqueous humor cytokines and chemokines in patients affected by uveal melanoma. Medicine 2020, 99, e22091. [Google Scholar] [CrossRef]
  203. Ghiam, B.K.; Xu, L.; Berry, J.L. Aqueous Humor Markers in Retinoblastoma, a Review. Transl. Vis. Sci. Technol. 2019, 8, 13. [Google Scholar] [CrossRef]
  204. Campanharo, C.V.; dos Santos Silveira, L.V.; Meira, D.D.; Casotti, M.C.; Altoé, L.S.C.; Louro, I.D.; Gonçalves, A.F.M.; Machado, A.M.; Paiva, B.S.; de Souza Inocencio, E.; et al. Pan-cancer and multiomics: Advanced strategies for diagnosis, prognosis, and therapy in the complex genetic and molecular universe of cancer. Clin. Transl. Oncol. 2024, 27, 2936–2954. [Google Scholar] [CrossRef] [PubMed]
  205. Sharma, A.; Debik, J.; Naume, B.; Ohnstad, H.O.; Sahlber, K.K.; Borgen, E.; Børresen-Dale, A.-L.; Engebråten, O.; Fritzman, B.; Garred, Ø.; et al. Comprehensive multi-omics analysis of breast cancer reveals distinct long-term prognostic subtypes. Oncogenesis 2024, 13, 22. [Google Scholar] [CrossRef] [PubMed]
  206. Rudnick, P.A.; Markey, S.P.; Roth, J.; Mirokhin, Y.; Yan, X.; Tchekhovskoi, D.V.; Edwards, N.J.; Thangudu, R.R.; Ketchum, K.A.; Kinsinger, C.R.; et al. A Description of the Clinical Proteomic Tumor Analysis Consortium (CPTAC) Common Data Analysis Pipeline. J. Proteome Res. 2016, 15, 1023–1032. [Google Scholar] [CrossRef] [PubMed]
  207. Chen, M.-J.M.; Li, J.; Wang, Y.; Akbani, R.; Lu, Y.; Mills, G.B.; Liang, H. TCPA v3.0: An Integrative Platform to Explore the Pan-Cancer Analysis of Functional Proteomic Data. Mol. Cell. Proteom. 2019, 18, S15–S25. [Google Scholar] [CrossRef]
  208. Geffen, Y.; Anand, S.; Akiyama, Y.; Yaron, T.M.; Song, Y.; Johnson, J.L.; Govindan, A.; Babur, Ö.; Li, Y.; Huntsman, E.; et al. Pan-cancer analysis of post-translational modifications reveals shared patterns of protein regulation. Cell 2023, 186, 3945–3967.e26. [Google Scholar] [CrossRef] [PubMed]
  209. Wang, X.; Zhao, J.; Marostica, E.; Yuan, W.; Jin, J.; Zhang, J.; Li, R.; Tang, H.; Wang, K.; Li, Y.; et al. A pathology foundation model for cancer diagnosis and prognosis prediction. Nature 2024, 634, 970–978. [Google Scholar] [CrossRef] [PubMed]
  210. Lu, M.; Zhan, X. The crucial role of multiomic approach in cancer research and clinically relevant outcomes. EPMA J. 2018, 9, 77–102. [Google Scholar] [CrossRef] [PubMed]
  211. van Delft, F.; Schuurbiers, M.; Muller, M.; Burgers, S.; Rossum, H.; Ijzerman, M.; Heuvel, M.; Koffijberg, H. Comparing modeling strategies combining changes in multiple serum tumor biomarkers for early prediction of immunotherapy non-response in non-small cell lung cancer. Tumor Biol. 2023, 46, S269–S281. [Google Scholar] [CrossRef]
  212. Maroco, J.; Silva, D.; Pina Rodrigues, A.; Guerreiro, M.; Santana, I.; Mendonça, A. Data mining methods in the prediction of Dementia: A real-data comparison of the accuracy, sensitivity and specificity of linear discriminant analysis, logistic regression, neural networks, support vector machines, classification trees and random forests. BMC Res. Notes 2011, 4, 299. [Google Scholar] [CrossRef]
  213. Moshawrab, M.; Adda, M.; Bouzouane, A.; Ibrahim, H.; Raad, A. Reviewing Federated Machine Learning and Its Use in Diseases Prediction. Sensors 2023, 23, 2112. [Google Scholar] [CrossRef]
  214. William, C.; Wangmo, C.; Ranjan, A. Unravelling the application of machine learning in cancer biomarker discovery. Cancer Insight 2023, 2, 1–8. [Google Scholar] [CrossRef]
  215. Qureshi, M.D.A.; Ramzan, M.F.; Amjad, F.; Haider, N. Artificial Intelligence in Metabolomics for Disease Profiling: A Machine Learning Approach to Biomarker Discovery. Indus J. Biosci. Res. 2024, 2, 87–96. [Google Scholar] [CrossRef]
  216. Wang, F.; Su, Q.; Li, C. Identidication of novel biomarkers in non-small cell lung cancer using machine learning. Sci. Rep. 2022, 12, 16693. [Google Scholar] [CrossRef]
  217. Kwon, H.-J.; Park, U.-H.; Goh, C.; Park, D.; Lim, Y.; Lee, I.; Do, W.-J.; Lee, K.; Kim, H.; Yun, S.-Y.; et al. Enhancing Lung Cancer Classification through Integration of Liquid Biopsy Multi-Omics Data with Machine Learning Techniques. Cancers 2023, 15, 4556. [Google Scholar] [CrossRef]
  218. Rigakos, G.; Razis, E.; Koliou, G.-A.; Oikonomopoulos, G.; Tsolaki, E.; Sperinde, J.; Chrisafi, S.; Zarkavelis, G.; Pazarli, E.; Batistatou, A.; et al. Evaluation of the Role of p95 HER2 Isoform in Trastuzumab Efficacy in Metastatic Breast Cancer. Anticancer. Res. 2021, 41, 1793–1802. [Google Scholar] [CrossRef] [PubMed]
  219. Goh, C.W.; Yang, B.; Chi, Y.; Wu, J. p95HER2 expression in HER2-positive breast cancer with primary resistance. Precis. Med. Sci. 2024, 13, 99–106. [Google Scholar] [CrossRef]
  220. Thapa, R.; Wilson, G.D. The Importance of CD44 as a Stem Cell Biomarker and Therapeutic Target in Cancer. Stem Cells Int. 2016, 2016, 2087204. [Google Scholar] [CrossRef]
  221. Thomas, D.; Sagar, S.; Liu, X.; Lee, H.-R.; Grunkemeyer, J.A.; Grandgenett, P.M.; Caffrey, T.; O’Connell, K.A.; Swanson, B.; Marcos-Silva, L.; et al. Isoforms of MUC16 activate oncogenic signaling through EGF receptors to enhance the progression of pancreatic cancer. Mol. Ther. 2021, 29, 1557–1571. [Google Scholar] [CrossRef]
  222. Duncan, K.D.; Pětrošová, H.; Lum, J.J.; Goodlett, D.R. Mass spectrometry imaging methods for visualizing tumor heterogeneity. Curr. Opin. Biotechnol. 2024, 86, 103068. [Google Scholar] [CrossRef]
  223. McGee, J.P.; Su, P.; Durbin, K.R.; Hollas, M.A.R.; Bateman, N.W.; Maxwell, G.L.; Conrads, T.P.; Fellers, R.T.; Melani, R.D.; Camarillo, J.M.; et al. Automated imaging and identification of proteoforms directly from ovarian cancer tissue. Nat. Commun. 2023, 14, 6478. [Google Scholar] [CrossRef]
  224. Li, M.; Zuo, J.; Yang, K.; Wang, P.; Zhou, S. Proteomics mining of cancer hallmarks on a single-cell resolution. Mass Spectrom. Rev. 2024, 43, 1019–1040. [Google Scholar] [CrossRef]
  225. Su, P.-R.; You, L.; Beerens, C.; Bezstarosti, K.; Demmers, J.; Pabst, M.; Kanaar, R.; Hsu, C.-C.; Chien, M.-P. Microscopy-based single-cell proteomic profiling reveals heterogeneity in DNA damage response dynamics. Cell Rep. Methods 2022, 2, 100237. [Google Scholar] [CrossRef] [PubMed]
  226. King, M.E.; Zhang, J.; Lin, J.Q.; Garza, K.Y.; DeHoog, R.J.; Feider, C.L.; Bensussan, A.; Sans, M.; Krieger, A.; Badal, S.; et al. Rapid diagnosis and tumor margin assessment during pancreatic cancer surgery with the MasSpec Pen technology. Proc. Natl. Acad. Sci. USA 2021, 118, e2104411118. [Google Scholar] [CrossRef]
  227. Sans, M.; Zhang, J.; Lin, J.Q.; Feider, C.L.; Giese, N.; Breen, M.T.; Sebastian, K.; Liu, J.; Sood, A.K.; Eberlin, L.S. Performance of the MasSpec Pen for Rapid Diagnosis of Ovarian Cancer. Clin. Chem. 2019, 65, 674–683. [Google Scholar] [CrossRef] [PubMed]
  228. Garza, K.Y.; King, M.E.; Nagi, C.; DeHoog, R.J.; Zhang, J.; Sans, M.; Krieger, A.; Feider, C.L.; Bensussan, A.V.; Keating, M.F.; et al. Intraoperative Evaluation of Breast Tissues During Breast Cancer Operations Using the MasSpec Pen. JAMA Netw. Open 2024, 7, e242684. [Google Scholar] [CrossRef] [PubMed]
  229. Pedrero, M.; Gamella, M.; Serafín, V. Chapter 19 Nanomachines and nanorobotics: Improving cancer diagnosis and therapy. In The Detection of Biomarkers; Ozkan, S.A., Bakirhan, N.K., Mollarasouli, F., Eds.; Academic Press: Cambridge, UK, 2022; pp. 503–543. [Google Scholar] [CrossRef]
  230. Syedmoradi, L.; Norton, M.L.; Omidfar, K. Point-of-care cancer diagnostic devices: From academic research to clinical translation. Talanta 2021, 225, 122002. [Google Scholar] [CrossRef]
  231. Fu, L.; Karimi-Maleh, H. Leveraging electrochemical sensors to improve efficiency of cancer detection. World J. Clin. Oncol. 2024, 15, 360–366. [Google Scholar] [CrossRef]
  232. Saasa, V.; Chibagidi, R.; Ipileng, K.; Feleni, U. Advances in cancer detection: A review on electrochemical biosensor technologies. Sens. Bio-Sens. Res. 2025, 49, 100826. [Google Scholar] [CrossRef]
  233. Kim, Y.J.; Rho, W.-Y.; Park, S.-m.; Jun, B.-H. Optical nanomaterial-based detection of biomarkers in liquid biopsy. J. Hematol. Oncol. 2024, 17, 10. [Google Scholar] [CrossRef]
  234. Campuzano, S.; Barderas, R.; Moreno-Casbas, M.T.; Almeida, Á.; Pingarrón, J.M. Pursuing precision in medicine and nutrition: The rise of electrochemical biosensing at the molecular level. Anal. Bioanal. Chem. 2024, 416, 2151–2172. [Google Scholar] [CrossRef]
  235. O’Brien, C.; Khor, C.K.; Ardalan, S.; Ignaszak, A. Multiplex electrochemical sensing platforms for the detection of breast cancer biomarkers. Front. Med. Technol. 2024, 6, 1360510. [Google Scholar] [CrossRef]
  236. Dosnon, L.; Rduch, T.; Meyer, C.; Herrmann, I. A Wearable In-Pad Diagnostic for the Detection of Disease Biomarkers in Menstruation Blood. Adv. Sci. 2025, 12, e05170. [Google Scholar] [CrossRef]
  237. Lee, J.S.; Lee, N.; Kashif, A.; Yang, S.-J.; Nam, A.R.; Song, I.-C.; Gong, S.-J.; Hong, M.; Kim, G.; Seok, P.; et al. S100A8 and S100A9 Promote Apoptosis of Chronic Eosinophilic Leukemia Cells. Front. Immunol. 2020, 11, 1258. [Google Scholar] [CrossRef] [PubMed]
  238. Baydar, E.; Celikkol, A.; GÜRdal, S.; Seber, S. Predictive Value of Serum Calprotectin Level in Response to Treatment, a New Inflammatory Marker in Patients with Breast Cancer Requesting Neoadjuvant Treatment. Namık Kemal Tıp Derg. 2023, 11, 12–16. [Google Scholar] [CrossRef]
  239. Argyris, P.P.; Slama, Z.M.; Ross, K.F.; Khammanivong, A.; Herzberg, M.C. Calprotectin and the Initiation and Progression of Head and Neck Cancer. J. Dent. Res. 2018, 97, 674–682. [Google Scholar] [CrossRef]
  240. Chen, L.; Shu, P.; Zhang, X.; Ye, S.; Tian, L.; Shen, S.; Ma, J.; Ai, F.; Li, X. S100A8-Mediated Inflammatory Signaling Drives Colorectal Cancer Progression via the CXCL5/CXCR2 Axis. J. Cancer 2024, 15, 3452–3465. [Google Scholar] [CrossRef] [PubMed]
  241. Wang, W.; Chang, J.; Jia, B.; Liu, J. The Blood Biomarkers of Thyroid Cancer. Cancer Manag. Res. 2020, 12, 5431–5438. [Google Scholar] [CrossRef]
  242. Wu, Z.; Jiang, D.; Huang, X.; Cai, M.; Yuan, K.; Huang, P. S100A8 as a Promising Biomarker and Oncogenic Immune Protein in the Tumor Microenvironment: An Integrative Pancancer Analysis. J. Oncol. 2022, 2022, 6947652. [Google Scholar] [CrossRef]
  243. Zhang, X.; Niu, M.; Li, T.; Wu, Y.; Gao, J.; Yi, M.; Wu, K. S100A8/A9 as a risk factor for breast cancer negatively regulated by DACH1. Biomark. Res. 2023, 11, 106. [Google Scholar] [CrossRef]
  244. Miller, P.; Kidwell, K.M.; Thomas, D.; Sabel, M.; Rae, J.M.; Hayes, D.F.; Hudson, B.I.; El-Ashry, D.; Lippman, M.E. Elevated S100A8 protein expression in breast cancer cells and breast tumor stroma is prognostic of poor disease outcome. Breast Cancer Res. Treat. 2017, 166, 85–94. [Google Scholar] [CrossRef]
  245. Hermani, A.; Hess, J.; De Servi, B.; Medunjanin, S.; Grobholz, R.; Trojan, L.; Angel, P.; Mayer, D. Calcium-Binding Proteins S100A8 and S100A9 as Novel Diagnostic Markers in Human Prostate Cancer. Clin. Cancer Res. 2005, 11, 5146–5152. [Google Scholar] [CrossRef] [PubMed]
  246. Ebbing, J.; Mathia, S.; Seibert, F.; Pagonas, N.; Bauer, F.; Erber, B.; Günzel, K.; Kilic, E.; Kempkensteffen, C.; Miller, K.; et al. Urinary calprotectin: A new diagnostic marker in urothelial carcinoma of the bladder. World J. Urol. 2013, 32, 1485–1492. [Google Scholar] [CrossRef] [PubMed]
  247. Xu, H.-r.; Pang, Z.; Wang, Q.-z.; Ou-Yang, S. S100A8 promotes the proliferation, migration and invasion in bladder cancer cells. J. Cancer 2025, 16, 1066–1077. [Google Scholar] [CrossRef]
  248. Miranda-Galvis, M.; Soares, C.C.; Carnielli, C.M.; Buttura, J.R.; de Sá, R.S.; Kaminagakura, E.; Marchi, F.A.; Leme, A.F.P.; Pinto, C.A.L.; Santos-Silva, A.R.; et al. New Insights into the Impact of Human Papillomavirus on Oral Cancer in Young Patients: Proteomic Approach Reveals a Novel Role for S100A8. Cells 2023, 12, 1323. [Google Scholar] [CrossRef]
  249. Shen, J.; Person, M.D.; Zhu, J.; Abbruzzese, J.L.; Li, D. Protein Expression Profiles in Pancreatic Adenocarcinoma Compared with Normal Pancreatic Tissue and Tissue Affected by Pancreatitis as Detected by Two-Dimensional Gel Electrophoresis and Mass Spectrometry. Cancer Res. 2004, 64, 9018–9026. [Google Scholar] [CrossRef] [PubMed]
  250. Kostakis, I.D.; Cholidou, K.G.; Kallianidis, K.; Perrea, D.; Antsaklis, A. The role of calprotectin in obstetrics and gynecology. Eur. J. Obstet. Gynecol. Reprod. Biol. 2010, 151, 3–9. [Google Scholar] [CrossRef]
  251. Walker, J.; Joy, A.A.; Vos, L.J.; Stenson, T.H.; Mackey, J.R.; Jovel, J.; Kao, D.; Madsen, K.L.; Wong, G.K.-S. Chemotherapy-induced weight gain in early-stage breast cancer: A prospective matched cohort study reveals associations with inflammation and gut dysbiosis. BMC Med. 2023, 21, 178. [Google Scholar] [CrossRef]
  252. Yasar, O.; Akcay, T.; Obek, C.; Turegun, F.A. Significance of S100A8, S100A9 and calprotectin levels in bladder cancer. Scand. J. Clin. Lab. Investig. 2017, 77, 437–441. [Google Scholar] [CrossRef]
  253. Sahin, Y.; Yucetas, U.; Ates, H.A.; Erkan, E.; Yucetas, E.; Temiz, M.Z.; Toktas, M.G.; Kadihasanoglu, M.; Topkaya, B.C. Improving the diagnosis of high grade and stage bladder cancer by detecting increased urinary calprotectin expression in tumor tissue and tumor-associated inflammatory response. Investig. Clin. Urol. 2019, 60, 343–350. [Google Scholar] [CrossRef]
  254. Ye, X.; Huai, J.; Ding, J. Diagnostic accuracy of fecal calprotectin for screening patients with colorectal cancer: A meta-analysis. Turk. J. Gastroenterol. Off. J. Turk. Soc. Gastroenterol. 2018, 29, 397–405. [Google Scholar] [CrossRef] [PubMed]
  255. Algeciras-Schimnich, A.; Preissner, C.; Theobald, J.; Finseth, M.; Grebe, S. Procalcitonin: A Marker for the Diagnosis and Follow-Up of Patients with Medullary Thyroid Carcinoma. J. Clin. Endocrinol. Metab. 2008, 94, 861–868. [Google Scholar] [CrossRef]
  256. Sun, J.; Chen, X.; Wang, Y. Comparison of the diagnostic value of CEA combined with OPN or DKK1 in non-small cell lung cancer. Oncol. Lett. 2020, 20, 3046–3052. [Google Scholar] [CrossRef]
  257. Sequist, L.V.; Skates, S.J.; Haas, W. A New Era of Protein-Based Assays for Cancer Early Detection. J. Thorac. Oncol. 2021, 16, 191–193. [Google Scholar] [CrossRef] [PubMed]
  258. Ostrin, E.J.; Bantis, L.E.; Wilson, D.O.; Patel, N.; Wang, R.; Kundnani, D.; Adams-Haduch, J.; Dennison, J.B.; Fahrmann, J.F.; Chiu, H.T.; et al. Contribution of a Blood-Based Protein Biomarker Panel to the Classification of Indeterminate Pulmonary Nodules. J. Thorac. Oncol. 2021, 16, 228–236. [Google Scholar] [CrossRef] [PubMed]
  259. Nolen, B.M.; Lomakin, A.; Marrangoni, A.; Velikokhatnaya, L.; Prosser, D.; Lokshin, A.E. Urinary Protein Biomarkers in the Early Detection of Lung Cancer. Cancer Prev. Res. 2015, 8, 111–119. [Google Scholar] [CrossRef] [PubMed]
  260. Lavoue, V.; Favier, A.; Franck, S.; Boutet, G.; Azuar, A.-S.; Brousse, S.; Golfier, F.; Uzan, C.; Vaysse, C.; Molière, S.; et al. French college of gynecologists and obstetricians (CNGOF) recommendations for clinical practice: Place of breast self-examination in screening strategies. Breast 2024, 75, 103619. [Google Scholar] [CrossRef] [PubMed]
  261. Vincent, Z.; Hornby, S.; Ball, S.; Sanders, G.; Ayling, R.M. Faecal calprotectin as a marker for oesophago-gastric cancer. Ann. Clin. Biochem. 2015, 52, 660–664. [Google Scholar] [CrossRef]
  262. Martinez-Garcia, E.; Lesur, A.; Devis, L.; Cabrera, S.; Matias-Guiu, X.; Hirschfeld, M.; Asberger, J.; van Oostrum, J.; Casares de Cal, M.d.l.Á.; Gómez-Tato, A.; et al. Targeted Proteomics Identifies Proteomic Signatures in Liquid Biopsies of the Endometrium to Diagnose Endometrial Cancer and Assist in the Prediction of the Optimal Surgical Treatment. Clin. Cancer Res. 2017, 23, 6458–6467. [Google Scholar] [CrossRef]
  263. Ngan, T.T.; Nguyen, N.T.Q.; Van Minh, H.; Donnelly, M.; O’Neill, C. Effectiveness of clinical breast examination as a ‘stand-alone’ screening modality: An overview of systematic reviews. BMC Cancer 2020, 20, 1070. [Google Scholar] [CrossRef]
  264. Fenton, J.J.; Rolnick, S.J.; Harris, E.L.; Barton, M.B.; Barlow, W.E.; Reisch, L.M.; Herrinton, L.J.; Geiger, A.M.; Fletcher, S.W.; Elmore, J.G. Specificity of Clinical Breast Examination in Community Practice. J. Gen. Intern. Med. 2007, 22, 332–337. [Google Scholar] [CrossRef][Green Version]
  265. Chen, F.K.; de Abreu, A.L.C.; Palmer, S.L. Utility of Ultrasound in the Diagnosis, Treatment, and Follow-up of Prostate Cancer: State of the Art. J. Nucl. Med. 2016, 57 (Suppl. S3), 13S–18S. [Google Scholar] [CrossRef] [PubMed]
  266. Woo, S.; Suh, C.H.; Kim, S.Y.; Cho, J.Y.; Kim, S.H. Diagnostic Performance of Prostate Imaging Reporting and Data System Version 2 for Detection of Prostate Cancer: A Systematic Review and Diagnostic Meta-analysis. Eur. Urol. 2017, 72, 177–188. [Google Scholar] [CrossRef]
Figure 1. Conventional vs. modern methods used in cancer detection and management. CT—computed tomography; CTCs—circulating tumor cells; EM—electron microscopy; EVs—extracellular vesicles; ICC—immunocytochemistry; IHC—immunohistochemistry; MRI—magnetic resonance imaging; MS—mass spectrometry; MSI—mass spectrometry imaging; PET-CT—positron emission tomography-computed tomography; PET-MRI—positron emission tomography-magnetic resonance imaging; US—ultrasonography; X-ray—radiography.
Figure 1. Conventional vs. modern methods used in cancer detection and management. CT—computed tomography; CTCs—circulating tumor cells; EM—electron microscopy; EVs—extracellular vesicles; ICC—immunocytochemistry; IHC—immunohistochemistry; MRI—magnetic resonance imaging; MS—mass spectrometry; MSI—mass spectrometry imaging; PET-CT—positron emission tomography-computed tomography; PET-MRI—positron emission tomography-magnetic resonance imaging; US—ultrasonography; X-ray—radiography.
Proteomes 13 00047 g001
Figure 2. Proteomic workflow used for protein biomarker detection in liquid biopsies. HPLC/UPLC—high-performance liquid chromatography/ultra-performance liquid chromatography; MS—mass spectrometry; MS/MS—tandem mass spectrometry; m/z—mass-to-charge ratio.
Figure 2. Proteomic workflow used for protein biomarker detection in liquid biopsies. HPLC/UPLC—high-performance liquid chromatography/ultra-performance liquid chromatography; MS—mass spectrometry; MS/MS—tandem mass spectrometry; m/z—mass-to-charge ratio.
Proteomes 13 00047 g002
Table 1. Histology vs. LB for cancer diagnosis, monitoring, and treatment.
Table 1. Histology vs. LB for cancer diagnosis, monitoring, and treatment.
FeaturesHistologyLB
Limitationslimited for certain types of cancerpotential for detection across multiple cancer types
limited to one or few biomarkers by multiplex detection in IHCpanel/multiple biomarkers assignment
time consuming, labor-intensivedetection of low-abundant proteins can be challenging; faster results
Invasivenessinvasivenon-invasive or minimally invasive
multiple biopsies are not feasible due to their invasive naturecan be performed more frequently with fewer risks to patient
Accuracyhigh specificity, depends on sample quality, interpretation variability and human errorhigh specificity and sensitivity, especially with advanced technologies
Risk to patientinfections, bleeding, pain, and prolonged recoveryminimal to no risk
Sample typetissue sample, usually from biopsy or surgical resectiondetect CTCs, DNA, EVs, proteins, metabolites from multiple tumor regions
Monitoring and fellow-upprovides a snapshot of the tumor at the time of biopsyallows for real-time monitoring of cancer
Molecular insightsinability to monitor dynamic changes, provides limited molecular insights, focusing on histological featuresdynamic insight, provides a broader range of molecular biomarkers
Early cancer detectionrisk of missing early or small lesions, limitation in detection of early-stage cancer or micrometastasesallows for early detection of cancer and minimal residual disease
Detection of tumor heterogeneitylimited; may not capture the clonal and molecular diversity within the tumorcan detect molecular variations across the entire tumor mass, including mutations and PTMs of proteins
Costgenerally highergenerally lower
Treatment decision-makingdelayed decision-making for treatmentaccelerate decision-making for treatment
Table 2. Computed tomography (CT) vs. LB for cancer diagnosis, monitoring, and treatment.
Table 2. Computed tomography (CT) vs. LB for cancer diagnosis, monitoring, and treatment.
FeaturesCTLB
Principleconsidered non-invasive, possible negative effects due to dose-cumulative effects of X-rays non-invasive/minimally invasive
Diagnostic abilityhigh-resolution images for detecting tumors, distant metastasis, and anatomical changesdetects molecular biomarkers such as mutations, gene expression, and tumor-related proteins
Sensitivityhigh sensitivity for detecting structural changes, particularly in large tumorsdepends on the biomarker used and the cancer stage but can detect early-stage cancers
Specificityvaries among tumor types, size, locationdepends on the biomarker panel
Tumor monitoringeffective for monitoring tumor size and response to treatmentmonitors continuously and non-invasive the response to therapy
Costscan be expensive, necessitates imaging equipmentless expensive than conventional imaging, the costs depends on the complexity of the test
Clinical applicationscommonly use in cancer detection, staging, monitoring, and evaluating metastasisused for early-stage cancer detection, monitoring minimal residual disease, and assessing treatment efficiency; earlier detection of metastasis at molecular level
Use in early detectiondetects tumors that have reached a certain sizedetects cancer even before clinical signs are present
Advantageshigh spatial resolution accuracy, effective for visualizing tumor anatomyprovides molecular insights into tumors, can be done repeatedly, and is non- or minimally invasive
Disadvantageslimited in early-stage cancer detection, X-ray exposure is disputable; not detects molecular characteristicsmany methods require additional validation, can have lower sensitivity for certain tumor types
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

Neagu, A.-N.; Bruno, P.S.; Josan, C.-L.; Waterman, N.; Morrissiey, H.; Njoku, V.T.; Darie, C.C. In Search of Ideal Solutions for Cancer Diagnosis: From Conventional Methods to Protein Biomarkers in Liquid Biopsy. Proteomes 2025, 13, 47. https://doi.org/10.3390/proteomes13040047

AMA Style

Neagu A-N, Bruno PS, Josan C-L, Waterman N, Morrissiey H, Njoku VT, Darie CC. In Search of Ideal Solutions for Cancer Diagnosis: From Conventional Methods to Protein Biomarkers in Liquid Biopsy. Proteomes. 2025; 13(4):47. https://doi.org/10.3390/proteomes13040047

Chicago/Turabian Style

Neagu, Anca-Narcisa, Pathea S. Bruno, Claudiu-Laurentiu Josan, Natalie Waterman, Hailey Morrissiey, Victor T. Njoku, and Costel C. Darie. 2025. "In Search of Ideal Solutions for Cancer Diagnosis: From Conventional Methods to Protein Biomarkers in Liquid Biopsy" Proteomes 13, no. 4: 47. https://doi.org/10.3390/proteomes13040047

APA Style

Neagu, A.-N., Bruno, P. S., Josan, C.-L., Waterman, N., Morrissiey, H., Njoku, V. T., & Darie, C. C. (2025). In Search of Ideal Solutions for Cancer Diagnosis: From Conventional Methods to Protein Biomarkers in Liquid Biopsy. Proteomes, 13(4), 47. https://doi.org/10.3390/proteomes13040047

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