Next Article in Journal
Gut Hormones and Inflammatory Bowel Disease
Previous Article in Journal
The Impact of the Microbiota on the Immune Response Modulation in Colorectal Cancer
Previous Article in Special Issue
The Role of Eosinophils, Eosinophil-Related Cytokines and AI in Predicting Immunotherapy Efficacy in NSCLC
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Future of Tumor Markers: Advancing Early Malignancy Detection Through Omics Technologies, Continuous Monitoring, and Personalized Reference Intervals

by
Irem Nur Savas
1 and
Abdurrahman Coskun
2,*
1
Department of Biochemistry and Molecular Biology, Institute of Health Sciences, Acibadem Mehmet Ali Aydinlar University, Istanbul 34752, Turkey
2
Department of Medical Biochemistry, School of Medicine, Acibadem Mehmet Ali Aydinlar University, Kayisdagi cad., No: 32, Istanbul 34752, Turkey
*
Author to whom correspondence should be addressed.
Biomolecules 2025, 15(7), 1011; https://doi.org/10.3390/biom15071011
Submission received: 8 May 2025 / Revised: 19 June 2025 / Accepted: 3 July 2025 / Published: 14 July 2025
(This article belongs to the Collection Feature Papers in Molecular Biomarkers)

Abstract

Malignant diseases represent a major global health challenge and are among the leading causes of death worldwide. Accurate early diagnosis is essential for improving outcomes and combating these conditions effectively. Currently, the diagnosis of malignancies relies heavily on radiological imaging and pathological examinations, which are often invasive and not cost-effective. As such, there is a growing need for non-invasive and accessible methods to detect cancer in its early stages. Tumor markers—biomolecules whose levels increase in malignancy and can be measured in blood or other biological tissues and fluids—offer a promising tool. However, the sensitivity and specificity of currently available tumor markers are insufficient for early detection, limiting their use primarily to disease monitoring rather than diagnosis. While ongoing research continues to identify novel tumor markers, the development of more effective early detection strategies requires more than the discovery of new biomarkers. The continuous monitoring of patients and individuals with a high tumor risk and the personalization of tumor marker interpretation are also critical. In this review, we (i) summarize the most commonly used tumor markers, (ii) examine strategies for developing novel biomarkers, particularly through omics technologies, (iii) explore the potential of continuous monitoring using wearable biosensors for early tumor detection, and (iv) discuss approaches to personalizing tumor marker interpretation to support early diagnosis and improve treatment outcomes.

Graphical Abstract

1. Introduction

Malignancies have caused profound devastation to human life, emotions, hopes, resources, and economies—few other conditions have had such a widespread and multifaceted impact. According to the World Health Organization, malignant diseases represent the second leading cause of death globally, following cardiovascular diseases [1]. The accurate diagnosis of malignancy constitutes the first and most critical step in combating cancer, because malignant cells tend to grow rapidly and metastasize to distant sites, making the disease significantly more difficult to treat at advanced stages. The doubling times of some common malignant diseases, presented in Table 1, highlight the urgency of early detection and underscore the narrow window available for initiating effective treatment. Therefore, early diagnosis is essential for effective management and improved patient outcomes. Currently, the diagnosis of malignant diseases primarily relies on radiological findings and the examination of patient tissue samples for signs of pathology. However, such approaches may be invasive and potentially harmful, posing significant challenges for broad implementation across the general population. As a result, many common malignancies—such as lung, breast, and gastrointestinal cancers—are often diagnosed at advanced stages, thereby reducing the likelihood of successful radical treatment [2,3,4]. To address this challenge and enable the detection of early malignancies, non-invasive approaches—such as the determination of tumor-specific biomolecules or circulating tumor cells (CTCs)—hold significant potential for early tumor discovery [5,6,7,8,9].
New strategies for the early detection of tumors should be cost-effective, non-invasive, and user-friendly, enabling their integration into routine clinical practice without causing significant discomfort or requiring extensive time. Although many tumor markers are widely employed in clinical practice, most of them lack specificity and their levels can be also elevated in non-malignant conditions (Table 2), often resulting in false positive findings and unnecessary biopsies [24]. Therefore, the identification of novel tumor-specific markers and the development of advanced algorithms to evaluate multiple markers simultaneously are crucial for detecting tumors at the earliest possible stage [25,26]. Given the variable doubling times of tumors (Table 1), effective monitoring is essential. The continuous surveillance of tumor-associated biomarkers may enable us to detect a malignancy at its earliest stage, offering a critical advantage for timely and effective treatment.
Tumor metabolism is also prone to inter-individual variances; therefore, genetic analysis is essential for selecting the most effective drugs or treatment strategies [43,44,45,46,47]. Both the interpretation of tumor marker levels and the assessment of treatment efficacy should be based on the individual’s clinical data rather than population means. In other words, a personalized approach is required—one that not only tailors the interpretation of tumor markers and treatment plans but also evaluates therapeutic effectiveness using the individual’s unique biological profile.
In this review, we (i) briefly overview the most commonly used tumor markers, (ii) present strategies for the development of novel markers, (iii) explore the potential of continuous monitoring to detect tumors at an early stage, and (iv) discuss how the interpretation of tumor markers can be personalized to facilitate early detection and thereby enable more effective treatment.

2. Tumor Markers

Tumor markers are biomolecules that are overproduced or structurally altered as a cause or consequence of malignant processes. Tumor markers can be found intracellularly or extracellularly, the latter are often released into the circulation. Many of them can also be detected in other body fluids such as interstitial fluid (ISF), urine, seminal fluid, tears, and saliva. Tumor-associated molecules exhibit wide structural variation—they are proteins (peptides, enzymes, transporters, and hormones), carbohydrates, or even lipids. Additionally, CTCs have gained attention for their diagnostic and prognostic potential in cancer. This review focuses primarily on tumor markers that can be detected in blood or other body fluids.

2.1. Proteins

The majority of tumor markers used in clinical practice are proteins. Although enzymes are proteins and several hormones are peptides or proteins, they are often classified separately due to their distinct biological functions. Proteins in use as tumor markers include oncofetal proteins such as alpha-fetoprotein (AFP) and carcinoembryonic antigen (CEA); protein fragments such as tissue polypeptide antigen and cytokeratin-19 fragment antigen 21-1 (CYFRA 21-1); and various immunoglobulins including Bence Jones proteins, as well as other proteins such as β2-microglobulin, chromogranin A, human epididymis protein 4 (HE4), thyroglobulin, squamous cell carcinoma antigen (SCC), and S100 protein (Figure 1A) [24].
Although the abovementioned markers are primarily used for the monitoring of cancer patients, a few have relatively higher diagnostic value, such as AFP and Bence Jones proteins [24]. Several proteins including ferritin, mesothelin, and bladder cancer–specific nuclear matrix protein 4 are considered potential tumor markers. However, their clinical utility as tumor markers is limited as they are still under investigation [48,49,50]. Additionally, certain cancer-associated membrane proteins may be released into the circulation through a proteolytic cleavage, and their soluble forms are also used as tumor markers, including sHER2 and sPD-L1 [51,52,53] (Figure 1A).
AFP and CEA are the most commonly used protein tumor markers and are briefly described in the following text.

2.1.1. Alpha-Fetoprotein (AFP)

AFP is an oncofetal protein and is the most widely used biomarker for hepatocellular carcinoma (HCC) [54]. AFP, an analog of albumin, is the predominant serum protein during the first trimester of gestation and declines to trace levels by 18 months after birth [55,56]. AFP serves as a carrier for various molecules, including retinoids and steroids, similarly to serum albumin. The serum AFP level in healthy adults is typically below 10 µg/L and elevated levels are primarily associated with HCC or non-seminomatous germ cell tumors [55]. Not many tumor markers are recommended for cancer screening and AFP is among them, alongside CA 125, human chorionic gonadotropin (hCG), and a few others [24]. AFP is used in combination with ultrasound for screening high-risk individuals with chronic hepatitis, including those infected with hepatitis B (HBV) or hepatitis C virus (HCV) or those with advanced liver fibrosis [24]. For early stage HCC, an AFP and ultrasound combination exhibits a detection rate of 63%, prompting the search for additional markers [57,58]. It seems that levels of AFP-L3 (an isoform of AFP) and des-gamma-carboxyprothrombin (DCP), also known as prothrombin induced by vitamin K absence-II (PIVKA-II), are not correlated with AFP levels. Hence, AFP-L3 and DCP may be elevated in HCC patients even when AFP remains within the normal range, supporting their role as complementary diagnostic markers [58,59]. In the most recent study [60], the diagnostic performance of a biomarker panel including AFP, AFP-L3, DCP, and CA 19-9 was evaluated in liver diseases. Receiver operating characteristic (ROC) curve analysis showed how the combination of the four markers outperformed each individual marker in distinguishing liver cancer from benign liver diseases (area under the ROC curve, AUC = 0.85) or from healthy controls (AUC = 0.95) [60]. More large-scale studies are needed to establish the role of AFP-L3 and DCP in clinical practice.

2.1.2. Carcinoembryonic Antigen (CEA)

CEA was initially identified in human colorectal cancer (CRC) [30]. CEA is an embryonal glycoprotein found in fetal tissues, primarily in the gastrointestinal tract [61]. The major role of CEA is in cell adhesion. Its production ceases before birth, resulting in only minimal levels in the blood of healthy adults (~3 ng/mL in non-smokers, ~5 ng/mL in smokers) [55]. Only 30–50% of CRC patients present with elevated CEA levels, so its use alone does not provide a reliable diagnosis [24]. However, after confirmed cancer diagnosis, CEA is a valuable biomarker in monitoring treatment response, detecting tumor recurrence, and guiding follow-up strategies [62,63]. More frequent CEA measurements in post-operative CRC patients contribute to overall survival, and it is recommended to repeat measurements every 3–6 months for 5 years [24]. Although primarily associated with CRC, elevated CEA levels can also be useful in the follow-up of gastric, lung, and breast cancer patients [24]. A considerable number of studies have reported that the inclusion of CEA in diagnostic panels comprising various markers increases their diagnostic accuracy [64,65,66,67,68,69,70]. As a supporting example, a panel consisting of CEA, CYFRA 21-1, neuron-specific enolase (NSE), SCC, and ProGRP demonstrated higher diagnostic sensitivity (88,5%) and specificity (82%) than each marker alone in detecting lung cancer in patients with symptoms suggestive of this cancer [70]. Therefore, despite its limited specificity, CEA remains an essential biomarker with significant clinical relevance.

2.2. Enzymes

In oncology, enzymes are generally used as non-specific tumor markers, since they reflect tissue damage rather than indicating a specific malignancy. As an exception to this, tissue-specific isoenzymes are more typical for certain tumors. For example, prostate-specific antigen (PSA) is specific to prostate tissue and is consequently used in the management of prostate cancer and NSE is highly specific for tumors of neuroendocrine origin (also tumors with neuroendocrine differentiation) [71,72]. Other enzyme tumor markers include lactate dehydrogenase, alkaline phosphatase, thymidine Kinase-1, and kallikrein-related peptidase 2 (Figure 1A) [73,74,75].
PSA stands out among all enzyme tumor markers due to its unique specificity for the prostate, and it is described below.

Prostate-Specific Antigen (PSA)

PSA, first isolated from prostate tissue [36], is a proteolytic enzyme belonging to the kallikrein serine protease family. Physiologically, PSA facilitates the liquefaction of seminal fluid by proteolytically cleaving gel-forming proteins, thereby enhancing sperm motility. When the prostatic epithelial tissue is disrupted, increased amounts of PSA are released into the circulation. This elevation may result from a prostate cancer, but can also occur due to benign prostatic diseases [76]. PSA is widely used for monitoring prostate cancer. Although a cut-off value of 4 ng/mL is useful in diagnostic evaluation, it does not provide sufficient sensitivity [76]. Various strategies have been developed to improve the diagnostic accuracy of PSA and to reduce the number of unnecessary biopsies (~60% of all cases) [77]. Initial strategies included PSA density (total PSA/prostate volume) measurements, and PSA density over 0.2 ng/mL/cm3 has been associated with a significantly increased risk of malignancy [78]. On the other hand, statistical prediction models that incorporate various molecular forms of PSA (such as free PSA, proPSA, and intact PSA) and kallikrein-related peptidase 2, along with patient age and digital rectal examination, have demonstrated enhanced diagnostic accuracy (up to an AUC of 0.8) compared to total PSA alone (AUC ~0.6). More recently, Kachuri et al. proposed adjusting PSA levels based on individual genetic variation [79]. In a large multi-ancestry study, they developed a PSA polygenic score that accounted for approximately 10% of PSA variability. Genetically adjusted PSA levels reduced unnecessary prostate biopsies by up to 31%, although further studies are needed to refine their clinical utility.

2.3. Hormones

Hormones, resulting from either excessive production by the original endocrine tissue or ectopic synthesis by non-endocrine tissues, can also serve as cancer biomarkers [55]. Calcitonin is the most frequently utilized hormone tumor marker (see below). Another hormone, hCG, primarily associated with germ cell tumors, has a specific clinical application in the screening of gestational trophoblastic neoplasia, a rare malignancy that may develop following a molar pregnancy [80]. Other hormone-based tumor markers include bombesin, prolactin, and adrenocorticotropic hormone; catecholamines, including homovanillic acid and vanillymandelic acid; and also gut hormones, such as vasoactive intestinal peptide and pancreatic polypeptide (Figure 1A) [24,55].

Calcitonin

The first association between the hormone calcitonin and cancer was established when elevated levels were detected in patients with medullary thyroid carcinoma (MTC) [33,81]. It is a polypeptide hormone primarily involved in calcium homeostasis and mainly produced by the thyroid gland as part of a larger precursor molecule procalcitonin (ProCT). In MTC patients, calcitonin has a well-established role in assessing prognosis and is also routinely used with CEA for pre- and post-operative follow-up. Calcitonin levels exceeding 50–100 ng/L have diagnostic potential for MTC [82]; however, calcitonin as a cancer biomarker has a limited value due to certain drawbacks. Although calcitonin levels in blood may discriminate between cancer and healthy populations, the primary issues are the absence of established cut-off values and the short half-life of calcitonin in serum (15–40 min). Moreover, calcitonin’s daily fluctuations pose an additional analytical constraint to its wider use as a cancer biomarker [81,82]. Studies focusing on determining age- and gender-specific cut-off values are expected to increase the diagnostic usefulness of calcitonin for MTC [83]. Fortunately, compared to calcitonin, ProCT exhibits a longer half-life (20–24 h), thus providing a greater analytical stability [82]. A meta-analysis reported that ProCT measurement provided high pooled sensitivity (90%) and specificity (100%) in detecting MTC, making it a promising alternative diagnostic marker [84]. However, no comparison between calcitonin and ProCT was included, and further studies are warranted to confirm the clinical utility of the latter [84].

2.4. Carbohydrate Antigens

Tumor-associated carbohydrate antigens are glycans covalently bound to proteins (glycoproteins) or lipids (glycolipids), which are expressed on the surface of the tumor cells [26,55]. While these structures are absent or present at very low levels on normal cells, they are often overexpressed or structurally altered in malignant cells, leading to aberrant glycan profiles. Tumor-associated carbohydrate antigens such as CA 19-9, CA 125, CA 15-3, CA 72-4, and CA 27-29 are predominantly found among mucins, which are heavily glycosylated carrier glycoproteins [26,55]. These antigens are shed into the circulation during malignant processes and their levels are used clinically in monitoring malignancies. Due to their generally limited specificity, the diagnostic use of these biomarkers often requires additional methods and is typically interpreted alongside imaging techniques. Among the commonly used carbohydrate antigens, CA 19-9, CA 15-3, and CA 125 are discussed below.

2.4.1. Carbohydrate Antigen 19-9

CA 19-9, also known as sialyl Lewis antigen A, is the best tumor marker available for monitoring pancreatic cancer [85]. The presence of the Lewis antigen is required for the biosynthesis of CA 19-9, therefore Lewis antigen-negative individuals (5–10% of the population) can produce minimal amounts of CA 19-9, which must be taken into consideration when evaluating the usefulness of this cancer biomarker. CA 19-9 has diagnostic utility in pancreatic cancer with a sensitivity of 79% and a specificity of 82% [86]. In the early stages of pancreatic cancer, the sensitivity has been reported as 76.1% [85]. Many strategies have been developed to increase the diagnostic potential of CA 19-9 [86,87]. One of the recommended approaches is the combination of CA 19-9 with other commonly used tumor markers, including CEA, CA 125, and CA 242, as well as novel markers such as macrophage inhibitory cytokine-1, mucin 5AC (MUC5AC) [86,87]. For instance, combining CA 19-9 with MUC5AC significantly enhances predictive diagnostic performance (AUC = 0.91) compared to CA 19-9 alone (AUC = 0.61) [88]. Another strategy involves assessing the genetic status of patients, specifically genes coding for proteins associated with CA 19-9 biosynthesis. Using genotype-dependent cut-off values based on Lewis-negative and secretor-negative status improved the sensitivity of CA 19-9 for detecting early stage pancreatic cancer from 76.1% to 87.2% [85]. Large-scale studies will be useful for supporting the clinical and diagnostic applications of CA 19-9.

2.4.2. Cancer Antigen 125

CA 125 is a well-established tumor marker that represents an epitope derived from the transmembrane mucin 16 (MUC 16) [89]. Elevated CA 125 levels (>35 U/mL) are most commonly associated with epithelial ovarian cancer, but they may also be observed in other malignancies as well as in various non-malignant conditions (Table 2). Although it is not recommended for routine screening in healthy individuals, CA 125 has potential in screening high-risk women, particularly those with a family history of ovarian and breast cancer, or women identified as BRCA1/2 mutation carriers [90]. Currently, CA 125 is the most effective serum marker for ovarian cancer, though HE4 demonstrates a comparable diagnostic sensitivity (75%) and specificity (90%) [90,91]. Nevertheless, considerable efforts are being made to identify novel biomarkers with an improved diagnostic performance [89]. A comprehensive study evaluated the diagnostic value of 92 potential biomarkers [91]. Among these, eight candidates with relatively high diagnostic potential were further assessed in combination with CA 125, and a CA 125 and ADAM8 protein combination yielded the best diagnostic performance. However, this combination resulted in only a modest improvement (2%) in diagnostic accuracy compared to CA 125 alone [91]. More recently, it has been reported that assessing the mutational status and glycosylation extent of MUC 16, alongside CA 125 measurements, may offer a more reliable tool for the early diagnosis of ovarian cancer [89].

2.4.3. Cancer Antigen 15-3

CA 15-3 is a glycoprotein fragment derived from MUC1, the latter being a protein product of the breast cancer-associated MUC1 gene. CA 15-3 is the most widely utilized serum biomarker for breast cancer [26,92]. Its primary clinical application is in the assessment of metastatic disease, where it is interpreted in conjunction with imaging and clinical findings [93]. Elevated levels of CA 15-3 (>40–50 U/mL) at cancer diagnosis have been associated with poor prognosis and poor survival outcomes, probably reflecting its ability to detect clinically silent micro-metastases [24,93,94,95]. However, in early stages, CA 15-3 levels generally remain within normal limits (≤30 U/mL), thereby limiting its effectiveness for early detection [24,93]. To sum up, as in many other types of cancer, a reliable tumor marker for early breast cancer detection is still lacking.
Recently, Sekacheva et al. reported that the combined use of the novel biomarker CA-62 with CA 15-3 significantly enhances diagnostic accuracy in breast cancer [96]. Unlike CA 15-3, CA-62 has been identified as an epithelial carcinoma marker associated with abnormal cell proliferation and has been elevated in early stage breast cancer. The diagnostic accuracy of CA-62 (97%) and CA 15-3 (40%) in stage I disease has been reported [96]. Another study evaluating the combined use of these two markers demonstrated 75% sensitivity and 100% specificity for stage I breast cancer, underscoring their complementary diagnostic potential [97]. Notably, while the diagnostic performance of CA-62 declines in advanced stages, that of CA 15-3 improves, suggesting that their simultaneous use may offer diagnostic benefit in breast cancer detection [96,97]. Nevertheless, further studies are required to validate the clinical applicability of the CA-62 + CA 15-3 diagnostic combination.

2.5. Circulating Tumor Cells (CTCs)

CTCs detach from the primary tumor or metastases and enter the circulation, thus contributing to metastasis [98]. Ever since their potential for early cancer diagnosis, as well as for prognosis assessment and metastasis monitoring, has been demonstrated, CTCs have been extensively investigated for their clinical applications in oncology [99,100,101,102,103]. In an early stage cancer, CTCs are rare in blood (<5 CTCs per 7.5 mL of blood), with a short half-life ranging from 1 to 2.4 h [104]. However, their concentration may serve as an informative tumor marker [98]. As an example, in non-small cell lung cancer (NSCLC), five CTCs in 7.5 mL of blood has been used as a baseline value, and more CTCs have been associated with a worse prognosis [105]. Another study reported that abnormal lung imaging results and a CTC value over 25 per 7.5 mL can distinguish lung cancer from benign lesions and has the potential to be used for screening purposes [106]. In breast cancer, five CTCs per 7.5 mL has been suggested as a cut-off for a metastatic stage, while one CTC/7.5 mL has usually been detected in the localized disease [107]. According to several large prospective studies and meta-analyses, patients with high CTC counts in the blood generally have an extremely poor prognosis [100,106]. In addition to the quantitative assessment of CTCs, an emerging application is their use in liquid biopsy, an innovative, non-invasive alternative to tissue biopsies. Liquid biopsies are a source of tumor-derived nucleotides or proteins, and can be used as a source for advanced omics analyses (see Section 3.1.3).

3. Strategies for Early Detection of Malignancies

Despite the availability of numerous tumor markers (Figure 1A), current clinical practice does not enable the reliable early detection of tumors using these markers. Therefore, new strategies are essential to identify and clinically validate novel tumor markers for routine application. These strategies should be built upon three key cornerstones: (i) the discovery and development of novel tumor markers through omics technologies, (ii) the continuous monitoring of individuals using wearable biosensors, and (iii) the personalization of individual data to facilitate tumor detection in an early stage, as detailed below.

3.1. Omics Technologies in Tumor Marker Discovery: Opportunities and Challenges in Malignancy Diagnosis

To detect malignancies at an early stage, the metabolic activities of tumor cells—which serve as traces indicating the presence of tumors—should first be identified. The basic structural units of living systems are biomolecules; therefore, the transformation of a normal cell into a malignant one is accompanied by changes in the activities of biomolecules or alterations in their abundance. During the malignant transformation of a cell, some silent genes may become activated, leading to an increased expression of specific proteins and/or elevated biosynthesis or degradation of other biomolecules such as oligosaccharides, lipids, and metabolites. This way, certain biomolecules may appear at much greater concentrations than in healthy individuals. It is not enough to discover novel tumor-specific biomolecules, they must be clinically validated. Unfortunately, despite the discovery of dozens of cancer-associated biomolecules, a small number of them have been clinically validated as tumor markers. Moreover, most of these were investigated before the advent of the omics era (Table 2).
The first tumor marker to be identified was the Bence Jones protein, discovered in 1847, which is still widely used today in the diagnosis of multiple myeloma [27,108]. In 1927, hCG was associated with pregnancy and only later was it recognized as a tumor marker for germ cell tumors and gestational trophoblastic disease [28]. These two proteins represent the first generation of tumor markers that appeared in the period from 1847 to 1960 [109]. Second-generation tumor markers (1960–1970) [109] emerged following a pivotal discovery of AFP in mice inoculated with liver cancer cells (in 1963) [29]. During these years, advancements in analytical technologies, most notably the development of the radioimmunoassay, enabled large-scale studies. In 1975, elevated serum CEA levels were detected in cancer patients [30]. Shortly thereafter, immunometric assays based on monoclonal antibody technologies greatly accelerated the discovery of tumor markers [24]. This progress led to the discovery of many carbohydrate antigens such as CA 125, CA 15-3, and CA 19-9, also known as the third generation of tumor markers (1970–1980), [109] which became essential tools in cancer management. The fourth generation of tumor markers (1980–1990) comprised genetic markers including BRCA1/2, HER2, and TP53, all of which had already been introduced into clinical practice [26]. Between 1990 and 2004, coinciding with the Human Genome Project, a significant number of oncogenes, proto-oncogenes, and tumor suppressor genes were identified [26]. Thus, for many years, tumor marker discovery efforts focused primarily on individual molecules, single genes, and specific molecular pathways. After 2005, the expansion of scientific knowledge and the acceleration of technological developments (such as high-throughput sequencing, micro-array technologies, quantitative mass spectrometry, and computational bioinformatics platforms) facilitated the emergence of omics technologies and shifted tumor marker research into a more comprehensive and innovative phase [26].
Ironically, despite great expectations, omics technologies have so far not yielded the anticipated tumor-specific and sensitive biomarkers suitable for early stage detection in clinical laboratories. However, this does not preclude the possibility that such biomarkers or panels thereof will eventually be identified, and there is hope that omics technologies will ultimately fulfill this promise. Fifth-generation tumor markers (2005–present) predominantly comprise omics-based biomarkers, which will be discussed in this section.

3.1.1. Application of Omics Approaches for Novel Tumor Marker Identification

The term “-omics” refers to the comprehensive analysis of various molecular components within biological systems [110]. From an oncological perspective, major omics technologies are used to gain a comprehensive understanding of tumor biology. Among them, genomics serves as a large-scale analysis of genes to identify genetic alterations driving tumor development. Transcriptomics is employed for the identification of tumor-specific gene expression profiles and the molecular classification of tumors. Proteomics is used to elucidate changes in the expression of thousands of proteins and, when possible, changes in protein functions resulting from tumor progression. Finally, metabolomics and lipidomics are employed to assess dynamic metabolic processes in cancer and to characterize distinct metabolic profiles influenced by a wide range of factors, including genetic alterations and environmental conditions [25,111,112]. Each of these approaches holds strong potential for improving early cancer diagnosis, whether by enabling the identification of tumor-specific genetic and molecular profiles or, more specifically, by facilitating the discovery of novel tumor markers.
In the omics-based approaches, the process of translating a newly discovered biomarker from initial identification to clinical application involves four main stages: discovery, analytical validation, an assessment of clinical utility, and clinical implementation, along with several intermediate steps [113]. These include precise disease characterization, sample collection, high-throughput data acquisition, data analysis, the identification of candidate biomarkers, independent validation, and determining the validity of the biomarker across large populations [113]. To date, many candidate tumor markers have been discovered and presented by utilizing advanced omics technologies (Figure 1B). Next-generation sequencing, which has accelerated the advancement of genomic technologies, has enabled the simultaneous detection of multiple genetic alterations and the identification of genetic profiles associated with specific tumors [114]. For instance, beyond the well-known BRCA mutations in breast cancer, alterations in genes such as BARD1, and pathogenic variants of RAD51C and RAD51D, reported to be associated with increased breast cancer risk, have also been identified [114]. On the other hand, TERT promoter mutations, which contribute to early tumorigenesis by promoting telomerase activation, have been associated with more than 50 malignancies through comprehensive genomic analyses [115,116,117,118]. Moreover, TERT-associated alterations manifest as elevated mRNA expression levels, and their potential as tumor biomarkers for early diagnosis and prognosis has been demonstrated across various cancers, including bladder cancer and glioma [115,116,117,118].
The availability of The Cancer Genome Atlas, Human Protein Atlas, and other large-scale databases enables in silico approaches for screening gene expression and protein profiles, identifying tumor-specific genetic and molecular signatures, and detecting novel biomarker candidates [119,120]. These platforms offer an alternative to conventional sample collection and experimental analysis. A large number of studies are currently being conducted using these data-driven strategies, with ongoing efforts to identify potential tumor biomarkers for possible use in diagnostic settings. As an example, Zalfa et al. identified a panel of 41 genes that were found to be expressed up to 300-fold higher in ovarian cancer compared to non-malignant samples, based on a database-driven transcriptomic analysis. Among them, a model consisting of eight genes (including ADGRG1, EPCAM, and ESRP1) could distinguish ovarian serous cystadenocarcinoma (n = 42) from healthy ovarian tissues (n = 3) with sensitivity and specificity reaching 100%. The RNA expression levels of these genes were confirmed in an independent sample set. Expanding the sample size may contribute to the further evaluation of their diagnostic potential [121].
Despite the vast amount of genetic and transcriptomic data that has already been collected, this information cannot be directly translated into protein-level expression. Proteomic studies, leveraging new technologies, have uncovered the early diagnostic potential of various proteins. For instance, many proteins, including osteopontin, midkine, galectin-3, and annexin A2, were initially discovered before the emergence of omics technologies, yet large-scale proteomic analyses later revealed their potential for the early detection of malignancy [122,123,124]. Elevated levels of osteopontin have been associated with poor prognosis across multiple malignancies, including breast, pancreatic, and bladder cancers. There were several reports of osteopontin exhibiting diagnostic sensitivity comparable or even superior to AFP in HCC [125,126]. However, similar to the traditional tumor markers, osteopontin can be elevated in certain benign conditions, thus posing a significant limitation [127,128]. Additionally, proteomic studies have facilitated the discovery of novel tumor markers, by identifying and/or quantifying differentially expressed proteins. For instance, cyclin-dependent kinase 3 and actin beta-like 2 have been presented as potential tumor markers detected in tissue samples [129], while apolipoprotein A2 [130], heatshock protein 90α [131], dickkopf-1 [132], dysbindin [133], MUC5AC [134], TIMP-1 [135], vitronectin, dermcidin [136], transthyretin, and angiotensinogen [137] have been identified in serum. Malignancies associated with the listed proteins are displayed in Figure 1B.
Although examples of such protein biomarkers can be substantially expanded across different cancer types, the overall diagnostic potential of these emerging markers rarely exceeds that of conventional biomarkers currently used in clinical practice. The main reason is that the superiority of novel protein biomarkers is typically demonstrated in small-scale studies with limited patient cohorts. In response to these limitations, a growing body of research has focused on the combined use of established biomarkers and newly identified candidates derived from omics-based analyses, aiming to improve diagnostic performance through complementary biomarker panels. As an example, Gao et al. identified, through proteomics, a novel bile biomarker, clusterin (CLU), to improve the diagnosis of cholangiocarcinoma. The reported sensitivity of CLU was 73.6% and its specificity was 90.1%. Subsequently, a seven-marker diagnostic panel was developed by combining CLU with serum CA 19-9 and five additional biochemical parameters, including indirect bilirubin and gamma-glutamyl transferase. This panel demonstrated improved diagnostic performance, achieving 90.3% sensitivity in an independent validation cohort [138]. To facilitate the clinical applicability of the panel, the researchers created a user-friendly online model that displays the diagnostic prediction with a percentage probability [138].
On the other hand, individual metabolomes reflect the individual conditions in a highly personalized and dynamic way, as they result from a wide variety of factors, such as genetics, comorbidities, and lifestyle. Tumor markers derived from metabolomic studies are typically organized into comprehensive panels comprising numerous metabolites, due to the inherent complexity and dynamic nature of metabolic pathways, which rarely allow a single metabolite to fully represent tumor-specific alterations. Nevertheless, even within panels, individual metabolites that enhance the potential to indicate tumor development have been identified in certain cancers. As an example, elevated levels of sarcosine have been associated with prostate cancer [139,140,141], while the phosphorylation and increased concentrations of carnitine-containing metabolites have been linked to breast cancer [114]. Among various tumor-associated metabolic alterations, elevated levels of 2-hydroxyglutarate, identified as an oncometabolite, have been observed in multiple cancer types, including glioma, multiple myeloma, and colorectal cancer [142].

3.1.2. Advancing Cancer Diagnostics Through Multi-Omics Integration

Analyses based on a single omics layer (such as genomics, transcriptomics, or proteomics) are often insufficient to reflect the molecular complexity of cancer. While genomic analyses are valuable for revealing an individual’s genetic predisposition, the functional consequences of such variants often remain unclear [110]. Similarly, a biomarker identified at the proteomic level may result from changes at several molecular layers, including genetic mutations, epigenetic modifications or metabolic derangements [110]. Therefore, by integrating multiple molecular dimensions, multi-omics approaches may provide a holistic framework for analyzing various molecular components and facilitate the identification of disease-specific biomarkers with improved sensitivity and specificity [104]. Although the benefits of multi-omics are increasingly acknowledged, the majority of studies focused on early cancer detection have integrated only two omics layers (such as genomics and epigenomics), falling short of fully leveraging multi-dimensional data [143]. Xu et al. integrated three omics layers, genomics, epigenomics, and transcriptomics, with a machine learning approach to differentiate ductal carcinoma in situ (DCIS) from breast cancer [144]. Since DCIS carries a risk of progression to invasive disease, reliable markers are needed for accurate early diagnosis [145]. In the study, gene expression analysis provided the highest classification accuracy, while the inclusion of DNA methylation and copy number variation data further increased the classification sensitivity. As a result, a 10-gene diagnostic panel was developed to distinguish DCIS from breast cancer with high accuracy, where the AUC was 0.99. The diagnostic potential of the panel was validated by RNA sequencing and transcriptomics using an independent external validation cohort [144].
Many novel biomarkers emerged through multi-omics technologies such as CDH3 [146] and INSM-1 [147] in thyroid cancer, DHFR and E2F3 [148] in neuroblastoma, and IQGAP1 [149] in gastric cancer. From a broader perspective, one initiative designed to maximize the benefits of multi-omics approaches is the development of Integrated Personal Omics Profiling (iPOP). This was designed for profiling an individual over time by integrating multi-omics data with clinical evaluations, using wearable devices and advanced molecular profiling, with the goal of developing predictive models for long-term health outcomes [150]. Although iPOP-generated data showed great potential to inform health and disease management [151,152], larger-scale studies are needed to confirm and expand these findings, including possible applications in the field of oncology.

3.1.3. Liquid Biopsy for Omics Analysis

Invasive tissue biopsies, commonly used for omics analyses, are not suited for cancer screening or early diagnostics [5]. The detection of CTCs, or the circulating products of tumors, such as fragments of nucleic acids or proteins in blood samples, provides a non-invasive alternative to conventional tissue biopsies [107]. This approach, referred to as a liquid biopsy, creates an opportunity for the implementation of omics analyses for screening, early cancer detection, and personalized omics profiling [143]. Liquid biopsy enables the non-invasive detection of tumor-specific mutations through the analysis of circulating free DNA (cfDNA). Examples of mutations detected via cfDNA are EGFR and KRAS, which are clinically important for the management of lung cancer [153,154], and mutations in TERT and TP53, which have broader applicability across various cancer types [118] (Figure 1B).
Another notable example is the detection of SEPT9 methylation in cfDNA, which has achieved clinical utility for the early detection of colorectal carcinoma [155]. CTC analysis has also been studied across a wide range of cancer types to date. Gu et al. have provided a comprehensive summary of CTC-associated tumor markers [101]. Recently, an attempt was made to identify a liquid biopsy signature (including CTCs, cfDNA, extracellular vesicles, circulating immune system, cf-nucleosomes, proteins, and microbiota) and generate a multi-marker panel in blood samples from patients with histologically confirmed pancreatic ductal adenocarcinoma to facilitate early detection as the first phase of a non-interventional prospective clinical trial [156]. On the other hand, several studies have presented multiple cancer screening tests based on omics studies of liquid biopsies [157,158,159,160]. These tests utilize various omics technologies, while leveraging traditional imaging and machine learning approaches to increase diagnostic accuracy, and aim to detect all cancer types while confined to their primary tissues [157,158,159,160]. As an example, Cohen et al. developed a liquid biopsy-based multi-cancer screening panel that integrates genetic mutations in cfDNA (across 1933 loci in 16 cancer-associated genes) with serum levels of eight protein biomarkers, including CA 125, CEA, and CA 19-9 [161]. The diagnostic sensitivity ranged from 91% to 98% for four cancer types (ovarian, liver, stomach, and pancreatic) while the specificity exceeded 99%. Notably, the sensitivity of the panel for stage I liver cancer was reported as 100%. The test yielded suboptimal results for breast and lung cancers. Nevertheless, the study highlights the potential utility of similar multi-analyte approaches, especially when optimized with carefully selected mutation targets and protein biomarkers tailored to specific cancer types [161]. Meanwhile, there is an increasing interest in the early diagnosis of lung cancer through liquid biopsies [162,163,164]. Mathios et al. performed genomic fragmentation analysis based on cfDNA analysis from liquid biopsy for the early diagnosis of high-risk lung cancer patients (e.g., with smoking history and pulmonary symptoms) [165]. Combining fragmentation profiles, clinical risk factors, CEA levels, and machine learning approaches, followed by CT imaging, 91% of patients with stage I/II lung cancer were detected with 80% specificity [165]. Moreover, liquid biopsies can be performed using advanced point-of-care biosensor platforms, enabling real-time monitoring and facilitating more dynamic, non-invasive cancer management strategies [166].

3.1.4. Integration of Omics Technologies and Artificial Intelligence

Studies conducted over many years have accumulated extensive data across multiple omics layers. Multi-omics approaches provide a comprehensive molecular landscape by enabling cross-validation across different omics layers. However, the integration of multi-omics data requires dedicated strategies to manage, harmonize, and effectively interpret diverse omics layers [167,168]. Moreover, developing multiplex panels composed of potential biomarkers is a valuable approach to achieving improved diagnostic performance, although it may also require more detailed and complex analyses [167,168]. Consequently, the need to process large-scale datasets and to develop diagnostic panels composed of biomarkers acting in synergy has created a demand for specialized algorithms. In today’s technological era, leveraging a wide range of artificial intelligence and machine learning approaches, such as support vector machines, random forest (RF), and deep neural networks, has become a promising and increasingly adopted strategy [169]. To facilitate multi-omics integration, various pathway and network-based systems, as well as hierarchical biological modules (e.g., deepKEGG), have been developed which leverage biological relationships between different molecular elements such as genes, microRNAs, and proteins [168,170]. On the other hand, the combination of multiple machine learning methods is frequently used as an effective strategy for selecting biomarker panels that enhance diagnostic performance [144,170,171]. As a result, inferential information has been derived from datasets that would likely be incomprehensible through manual analysis alone.
To date, various omics-based studies supported by artificial intelligence have been carried out for diagnostic purposes. These include the identification of novel miRNA signatures related to renal cell carcinoma [172], transcriptomic biomarkers for the diagnosis of NSCLC and HCC [173,174], and epigenomics profiles distinguishing malignant and benign tumors of the central nervous system [175]. Tao et al. developed a machine learning-based statistical model using cfDNA-based genome sequencing to improve the detection accuracy of early-stage HCC. The model, based on an RF algorithm adapted for whole-genome sequencing analysis (a novel weighted RF-driver model), achieved a maximum predictive performance with an AUC of 0.920 in independent validation cohorts [176]. Commercial cancer diagnostic and screening initiatives have also employed machine learning methods and yielded promising results [161,177]. Among these, a blood-based test that integrates cfDNA and protein biomarkers and applies machine learning—including convolutional neural networks—has been evaluated and validated in large-scale clinical trials for early CRC detection [178,179]. Accordingly, the data obtained from the PREEMPT CRC® trial (NCT04369053), which included 32,731 participants across more than 200 centers in an average-risk population, demonstrated a sensitivity of 79.2% for CRC and a specificity of 91.5% for advanced colorectal neoplasia [177].
Recently, Nagarkar et al. developed a multi-cancer early detection test based on the serum metabolome with the aid of machine learning [180]. Through untargeted serum metabolite profiling by high-resolution mass spectrometry, followed by selective filtering steps, a panel of 2709 metabolites was identified. The researchers listed 100 specific metabolites that contributed the most to diagnostic accuracy. When implemented, the panel was able to detect 30 cancer types, including early stages, with a sensitivity of 90–100% and a specificity of 99.2%. For example, the detection sensitivity and specificity for Stage I–II ovarian cancer were reported to be 100% and 96%, respectively [180].
As discussed above, since the early 2000s, significant efforts and extensive research have been undertaken to enable early cancer detection through the use of omics technologies. Numerous candidate tumor biomarkers have been rapidly discovered and reported in preclinical studies; however, translating these biomarkers into clinical applications requires large-scale validation efforts that follow a longer timeline. Many biomarkers reported to exhibit high performance were initially identified in small, single-center studies with limited patient populations, and are often subject to reproducibility limitations. For example, a recent study with approximately 12 years of follow-up investigated the association between 1463 plasma proteins and the risk of 19 different cancers, including CRC [181]. The authors reported that they were unable to replicate previously identified protein–risk associations, particularly those found in small CRC cohorts (n ≤ 100) [182,183]. Different sampling procedures and different analytical methodologies result in inconsistencies among study findings. Overcoming these challenges will require the implementation of standardized study protocols. On the other hand, the increasing volume and complexity of data, along with the growing number of studies and candidate biomarkers, have benefitted considerably from advances in bioinformatics and artificial intelligence, which have facilitated the organization, analysis, and interpretation of multidimensional datasets. The integration of artificial intelligence may provide the necessary support to unlock the long-anticipated potential of advanced omics technologies. However, the technologies and the resulting data are quite heterogeneous, and sophisticated methodologies are still required to properly integrate them and ensure clinical applicability [184]. Ongoing efforts are focused on validating promising single biomarkers, multi-marker panels, and molecular profiles across large patient cohorts. Feasible clinical translation requires reproducibility in independent research groups, standardized detection technologies, accessibility, and validated improvements in both accuracy and cost-effectiveness compared to existing diagnostic practices.

3.2. Continuous Monitoring of Malignancies Using Wearable Biosensors for Tumor Markers

Considering cancer’s major global impact on health, monitoring techniques for early cancer diagnosis have consistently attracted significant interest. Although traditional diagnostic methods such as imaging and biopsies are essential in the current state of healthcare technologies for cancer, they have fundamental limitations, including healthcare professional and laboratory resource requirements, the invasiveness of procedures, and high costs. The requirement for patients to visit specialized laboratories for measurements limits accessibility and the feasible testing frequency for screening, early diagnosis, and monitoring—particularly in high-risk individuals and those with rapidly progressing malignancies characterized by short tumor doubling times (Table 1), where more frequent assessments are critical [185]. Therefore, there is a growing demand for innovative technological approaches in the digital era. The development of wearable biosensors based on non-invasive sampling for the continuous measurement of tumor markers may provide effective clinical monitoring in oncology.
In recent years, wearable sensor technologies have been significantly improved and are now actively used in health and disease management [186,187,188]. These devices can analyze various body fluids, including ISF, seminal fluid, sweat, urine, tears, saliva, and breath [189]. Biosensors that are directly placed on the body, such as patches or electronic tattoos, integrated into clothing, or incorporated into accessories like smartwatches, rings, and bandages, collect and analyze personal physiological and activity data [188,189]. The wireless transmission of this data to the smart devices of users and healthcare providers may facilitate remote monitoring. While continuous measurements and real-time health feedback via wearable biosensors have become widespread for the elderly, individuals with disabilities, and chronic disease management, its application in oncology remains limited [190,191]. To the best of our knowledge, wearable biosensors capable of continuously measuring tumor markers in body fluids are not yet available; however, significant progress has been made, leading to the development of fully portable platforms [192] (Table 3).
With today’s advanced technology, tumor marker biosensors can perform rapid and sensitive on-site measurements using miniature devices. In the early stages of tumor development, due to the localized settlement of cancer cells, tumor marker concentrations in blood remain low, while levels in biological fluids such as sweat, tears, and ISF are typically even lower [224,225]. The detection limits of conventional diagnostic techniques are typically too high to detect these biomarkers at such minute levels. However, these can be effectively detected via biosensors. Biosensors employ various measurement technologies, including optical, colorimetric, piezoelectric, and electrochemical methods, which have been comprehensively described in several reviews [226,227]. Conductive materials used in portable biosensors such as MXenes [228], metal-organic frameworks [229], quantum dots [193], and conductive polymers [230] exhibit enhanced sensitivity, thus enabling the identification of tumor markers at otherwise undetectable levels. Biosensors can detect tumor markers, including CEA, AFP, PSA, CA 19-9, and CA 125 at ultra-low concentrations, e.g., pg/mL or even fg/mL [231,232].
Moreover, biosensors enable a rapid sample analysis, i.e., an instantaneous assessment of biomarker levels [233], particularly electrochemical biosensors, which show a response time of less than 5 min for CEA [234] and PSA [235], and even 35 s for AFP [202].
Biosensor technologies based on the sampling of blood and other body fluids are discussed below.

3.2.1. Biosensors for Tumor Biomarker Analysis from Blood

Since a great majority of clinically validated tumor biomarkers originate from the blood (Table 2), most biosensors are designed for the blood-based samples. Today, biosensors for nearly all common blood tumor markers are available; however, many still require laboratory-based set-ups [227,236]. Advances in miniaturized materials offer the potential for adaptation to point-of-care applications. Currently, portable sensors developed for fully integrated on-site use, and capable of operation with small table-top set-ups, are exemplified in Table 3. Advanced biosensors are now available for various applications, featuring sophisticated operating systems, integrated memory, wireless network connectivity, and the capability to provide real-time feedback to smartphones [237]. He et al. described a polymer-based compact and portable immunosensor for PSA [205] (Figure 2). The electrode surface was supported by gold film for enhanced specificity, and air plasma treatment and graphene were used for increased sensitivity where LOD was 0.38 fg/mL. Results are monitored via a synchronized smartphone, thus facilitating the early diagnosis of prostate cancer. In another study by Ibanez-Redin et al., a flexible electrochemical immunosensor was presented for CA 19–9 measurement (LOD: 0.07 U/mL) in serum and cell lysates by coating screen-printed carbon electrodes with carbon black and polyelectrolytes [197]. This biosensor is suitable for large-scale production and disposable use and offers the potential for frequent and easy CA 19-9 measurements in pancreatic cancer patients.
AFP is routinely measured by immunological assays (Table 2). As an alternative, various portable biosensors were developed for AFP detection [202,203,211,238]. A very recent study introduced an electrochemical immunosensor based on a nanocomposite composed of toluidine blue, gold nanoparticles, Fe3O4, and reduced graphene oxide (rGO) [202]. This immunosensor can detect serum AFP in just 35 s, demonstrating high concordance with ELISA-measured results in clinical serum samples (R2 = 0.998). The system equipped with a simple operating system was proposed for liver cancer screening at the point of care [202]. Chu et al. [211] developed silk-based and flexible sensors for detecting AFP and CEA biomarkers in blood. Photonic crystal barcodes provide excellent color properties and high-sensitivity measurements for fluorescence-based analysis (with a detection range of 10–1000 ng/mL for both AFP and CEA), while the graphene-based microcircuit system also functions as a motion sensor [211]. Researchers have also incorporated multilayered microchannels into biosensors for wearable applications, providing a fluidic pathway for sample transport and detection. Since the sensor currently uses a fingertip blood sample, integrating microneedles into the system may allow its adaptation for wearable use [211].
A recent comprehensive review of Foroozandeh et al. covered analytical features of biosensors for ovarian cancer diagnosis [239]. Among these, Srilikhit et al. developed a point-of-care immunosensor for the simultaneous detection of CA 125 (LOD: 0.6 U/mL) and CEA (LOD: 0.15 ng/mL) based on whole blood sampling [206]. By integrating a plasma separation membrane within a fluidic cell (Flu-iDCE) fabricated from an acrylic sheet, the system enables the direct analysis of whole blood, eliminating the need for centrifugation, thus offering a simplified sample preparation process. This innovation provides greater accessibility and operational convenience for the rapid and efficient detection of CA 125 and CEA [206]. In another example, a screen-printed electrochemical biosensor with multi-walled carbon nanotubes/thionine/gold nanoparticles for the detection of CA 125 transmits the measurement results to smartphones via Bluetooth. The device, which analyzes serum samples, also stands out with its low limit of detection (2 mU/mL) [200].
Biosensors developed for CTCs are also becoming popular in early cancer diagnosis [232]. Various immunosensors have been designed to detect and isolate CTCs, which are present in extremely low concentrations in circulation (see Section 2.5. Circulating Tumor Cells), especially in early stages. Among these, several methods utilizing immunoaffinity and immunomagnetic bead-based approaches, enhanced with micro and nanostructures to improve CTC detection efficiency, have been commercialized [240,241]. However, due to the differences between CTC detection technologies and the lack of a standardized method, diagnostic or prognostic evaluation criteria currently depend on the measurement method [107].
Biosensors based on blood sampling are not entirely suitable for continuous or frequent monitoring. However, for many biosensors that have not yet been standardized for clinical use, using blood as a sample enables their performance to be validated by comparison with conventional diagnostic tests [227].
Alternatively, biomarker measurements based on easily accessible body fluids are considered more feasible approaches for non-invasive, continuous monitoring.

3.2.2. Biosensors for Tumor Biomarker Analysis from Other Body Fluids

Early cancer detection efforts are increasingly focused on non-invasive technologies that enable continuous monitoring. Wearable or portable sensors that can analyze easily accessible body fluids such as sweat, saliva, urine, tears, and ISF offer suitable platforms for more frequent measurements. However, clinical application of these fluids for early diagnosis requires validated cancer biomarkers and standardized measurement methods [242]. Biosensors developed for the detection of tumor markers in various body fluids are summarized below.
ISF is the body fluid that surrounds tissue cells and has a composition quite similar to blood plasma [243]. Tumor markers such as AFP, CEA, and CA 15-3 can be detected in ISF [244]. For example, ISF collected from breast tissue offers great detection potential for various biomarkers in early stage breast cancer. Huang et al. developed a microneedle patch that enables minimally invasive and rapid extraction of ISF (~1.29 µL ISF in 1 min) from breast tissue. The patch was fabricated by UV-induced polymerization and cross-linking of acrylic acid with gelatin methacrylate, which enables the efficient removal of the sample. This approach offers the potential for earlier breast cancer detection by analyzing tumor markers, CEA and CA 15-3, in ISF, compared to traditional blood tests and imaging methods such as micro-CT and ultrasound (Figure 3A) [245]. Tumor-derived exosomes can also be used as a biomarker for early tumor identification [246]. Park et al. proposed an ISF-based biosensor for measuring glypican-1-positive exosomes for CRC diagnosis. The developed sensor can facilitate early tumor detection by non-invasive means, but additional processing is needed for application in wearable form [247].
Saliva is another easily accessible body fluid enriched in numerous tumor-associated biomarkers. Various biosensors have been developed for the saliva-based measurement of tumor markers, including PSA, CA 125, CA 72-4, CA 19-9, CEA, CYFRA 21-1, p53, TNF-α, IL-1β, and matrix metalloproteinase-9 [248,249,250]. In the study by Bratei et al., 3D needle-type stochastic microsensors were employed for the simultaneous and highly reliable analysis of CA 72-4, CA 19-9, CA 125, and CEA in biological fluids, including saliva. A key advantage of the method is its independence from the complexity of biological samples, ensuring accurate and reliable detection. The innovative sensor design incorporated boron- and nitrogen-decorated graphene, enhancing conductivity and stability [248]. The sensor enabled biomarker analysis with a broad linear range and low detection limits, establishing it as an efficient tool for early cancer screening [248]. Previously, CYFRA 21-1 was proposed as a potential biomarker for oral cancer [250,251]. Various biosensors have been developed for CYFRA 21-1 analysis based on saliva sampling [216,217,252,253]. Among them, Tofighi et al. proposed a portable silver nano-ink-assisted paper immunosensor for enhanced conductivity and stability. Electrochemical measurements provide a wide measurement range (0.0025–10 ng/mL) [216]. In the study, unprocessed real human saliva samples from an early stage oral cancer patient were used; however, the applicability of salivary CYFRA 21-1 as a biomarker for other cancers was not evaluated.
Urine has been used for many years for the detection of various diseases including cancer, due to its non-invasive collection and the presence of a wide range of biological markers.
Biosensors designed for urine analysis generally operate by using small (1 cm) microfluidic/disposable chip-structured sensors [219]. Khan et al. comprehensively listed over 100 urine-based biomarkers associated with various cancers, including lung, breast, colorectal, bladder, and prostate cancer [232]. Among these, biomarkers that can be measured using urinary biosensors are currently limited. For instance, in prostate cancer diagnosis, various sensors have been developed for the detection of volatile organic compounds, potential markers such as glypican-1, annexin A3, and endoglin, as well as tumor-derived DNA and RNA [254]. However, a major limitation remains the lack of validation through large-scale studies. In another example, Yang et al. developed an indium gallium zinc oxide-field effect transistor-based biosensor integrated with a machine learning algorithm for detecting bladder cancer in clinical urine samples [219]. In addition to nuclear matrix protein 22, the analysis of protein biomarkers, carbonic anhydrase 9, cluster of differentiation 47, cytokeratin 18, and cytokeratin 8 was proposed for more accurate bladder cancer diagnosis. The combined analysis of the above-listed five biomarkers achieved a detection accuracy of 95.0%. The system, which transmits analysis results via a wireless Bluetooth unit, was presented as a potential candidate for clinical applications in bladder cancer diagnosis [219].
Sweat, as a body fluid with a rich composition and easy accessibility, has shown a rising trend in biomarker analysis. Numerous wearable biosensors have already been developed for the detection of various biomarkers, including proteins, metabolites, and electrolytes [189]. However, sweat-based measurements of cancer-related biomarkers have not followed a similar trend, and studies in this area remain quite limited. CEA can also be detected in sweat. A wearable electrochemical biosensor was recently developed for rapid and quantitative testing of CEA (with a linear range of 0.2–100 ng/mL). The system employed a microfluidic sweat collector for sampling and AuNPs/rGO-modified screen-printed electrodes for electrochemical analysis [255]. However, additional studies are needed to evaluate its clinical significance.
Tears contain various molecules with the potential to reflect the physiological state of the body [256]. Studies have demonstrated the potential of tears for the early diagnosis and monitoring of various cancers [257,258]. Elevated levels of lactoferrin and cystatin C have been observed in eye cancer, while cystatin SA, lacryglobin, and miRNAs (miR-21 and miR-200c) have been found at higher concentrations in the tears of breast cancer patients [256,259]. Additionally, lacryglobin levels are elevated in the tears of patients with colon, prostate, and lung cancers [257]. An innovative approach involves the use of tear fluid for the early detection of breast cancer through a plasmonic SERS platform equipped with a gold-decorated, hexagonal-close-packed polystyrene (Au/HCP-PS) nanosphere monolayer, enabling femtomole-scale detection [220]. The measurements are based on the Raman scattering profiles of various biomarkers, including collagen, carotenoid, tyrosine, and phenylalanine. This technology reportedly achieved a detection sensitivity of 92% and specificity of 100% for breast cancer [220] (Figure 3B). While tear sampling is non-invasive and can be conducted using absorbent materials or micro-capillary tubes, it presents challenges such as the small sample volume, evaporation issues, and the impact of volumetric changes in tear production on the concentration of its biomolecules.
Exhaled breath contains thousands of distinct organic molecules, and the profile of volatile organic compounds (VOCs) varies between healthy and diseased states [260]. The early and non-invasive detection of lung cancer represents one of the most compelling potentials of breath analysis [261]. Wearable and portable devices designed as electronic noses have attracted significant attention for exhalation-based analysis [260,262]. These devices operate by detecting various VOCs, such as benzaldehyde, 2-ethylhexanol, isopropanol, and n-butanol, which are elevated in lung cancer [263,264]. Recently, Zompanti et al. developed a system in which exhaled breath is collected on absorbent cartridges and subsequently analyzed via a portable device [265]. This system was clinically evaluated to assess changes in the breath-print before and after the surgical resection of lung cancer, and was reported to detect recurrence with 91% accuracy [265]. Portable biosensors able to detect colorectal and prostate cancer from exhaled breath samples have also been developed [266,267]. Since current portable devices for cancer detection are relatively bulky, research is underway to develop miniaturized systems which would be user-friendly, and linked to smartphones [263]. The establishment of standardized protocols holds promise for enabling real-time, non-invasive diagnosis and monitoring of cancer patients.
Recent biosensors have significantly improved by offering a broad linear range, a high sensitivity, and a low detection limit, thus enabling biomarker detection in early stages [232]. Despite the considerable advancement in analytical capabilities [268], the integration of biosensors for tumor marker detection into fully integrated wearable platforms currently lags considerably behind other existing wearable sensor applications.
For instance, non-invasive wearable glucose sensors based on ISF sampling are in use for diabetes management [269]. In addition, researchers have developed a variety of wearable biosensors, including a ring-format device for the sweat-based analysis of female reproductive hormones [270], patch-format sensors for levodopa monitoring in Parkinson’s disease management [271,272,273], sweat-based metabolite detection for dietary applications [274], and also an ultrasound breast patch that provides deep tissue scanning for breast cancer monitoring [275]. Numerous modifications of biosensor technology could be described, reflecting the expanding scope and capabilities of wearable biosensors [189]. Considering the advanced biosensors developed for tumor marker measurement, the integration of non-invasive sampling modules—such as microfluidic technologies—and the use of biocompatible materials that provide sufficient softness and flexibility for on-skin applications would enable the transition of these biosensors into wearable platforms suitable for continuous monitoring.
Biosensor applications still face technical challenges such as ensuring stability, standardization, and reproducibility, as well as dealing with rapid degradation, cross-reactivity, and non-specific interference. The complexity of biological samples can affect detection performance, and structural similarity between molecules may cause false positives. Reliable performance requires strategies that reduce non-specific measurements [276]. Additionally, the detection of most cancer biomarkers requires high-affinity sensors, which is often achieved by aptamers and antibodies [232,239]. Biosensors utilizing natural biological recognition elements often exhibit limited functional stability during continuous use and are sensitive to environmental factors such as temperature fluctuations, which may compromise their long-term performance. However, as an alternative to natural recognition elements, molecularly imprinted polymers, which are resistant to degradation and have long-term stability, have the potential to overcome this limitation by enabling highly selective continuous measurements [223,274,277,278]. In addition, achieving consistent and comparable results with high reproducibility can be challenging across different biosensor platforms developed through various manufacturing processes. To address this, it is important to establish standardized fabrication protocols, measurement procedures, and validated performance criteria to ensure cross-platform compatibility and promote broader clinical and commercial adoption [276].
Nonetheless, elevated levels of tumor markers in non-malignant conditions remain key challenges, leading to diagnostic uncertainty. Advances in wearable biosensor technologies that allow the continuous measurement of tumor markers, along with other developments involving the identification and personalization of cancer-specific tumor markers, are likely to facilitate early stage cancer diagnosis.

3.3. Personalization of Tumor Markers

The interpretation of laboratory data, including tumor markers, relies on a comparison with the reference data, so the latter must be reliable. Despite considerable variance across different individuals having the same tumor, common reference intervals (RIs) are still widely used to interpret tumor marker levels. It should be noted that RIs are derived from population data and reflect population-level characteristics rather than individual-specific values [279,280]. Consequently, interpreting levels of tumor markers relying on the population-derived RIs may lead to misinterpretation. Hence, a result considered normal at the population level may, in fact, be abnormal for an individual, and vice versa.
The concentration of biomolecules, including tumor markers, fluctuates around a set point [281]. The range of this fluctuation can be determined using longitudinal data from a group of individuals referred to as within-subject biological variation (CVI), which reflects population-level estimates. Alternatively, we can use the individual’s own data, known as within-person biological variation (CVP), which is specific to that individual. Finally, the variation in the analyte levels among different individuals is known as between-subject biological variation (CVG) [282]. The ratio of CVI to CVG is known as the index of individuality (II). For a biomarker with an II less than 0.6, marked individuality is assumed, and population-based reference intervals (popRIs) are not recommended for interpreting patient results. Conversely, if the II is greater than 1.4, population-based reference intervals are considered appropriate for interpreting measurement results [283]. As shown in Table 1, the majority of tumor markers used in clinical practice exhibit marked individuality, indicating that although popRIs are currently used for interpretation, they are not suitable for these markers. Therefore, it is essential to develop new algorithms to estimate person-specific reference intervals—i.e., personalized reference intervals (prRIs)—as well as algorithms to monitor patients based on their own data rather than population means.

3.3.1. Personalized Reference Intervals for Tumor Markers: A Precision Medicine Approach

An analyte RI has upper and lower limits, thus representing the fluctuation of that analytes around a homeostatic set point (HSP). A prRI is estimated using an individual’s own data. Metabolically, the fluctuation of an analyte corresponds to its CVP. Both the HSP and the variation around it can be calculated using serial measurements of analyte concentrations in samples taken at the same time of day over days, weeks, or months, as detailed below.
H S P = x 1 + x 2 + x 3 + + x n n
where xi is the result of the ith measurements and n is the number of samples included in the calculation.
The variation around the HSP is the Gaussian combination of the CVP and the variation introduced by the measurement procedure, which can be considered as analytical variation (CVA), as formulated below:
T V = t α / 2 × C V P 2 + C V A 2
where tα/2 is the t-distribution value corresponding to n − 1 degrees of freedom.
The prRI can be considered as the prediction interval for the next measurement and can be calculated as shown below [284,285,286,287].
p r R I = H S P t α / 2 × n + 1 n ( C V P 2 + C V A 2 )
The use of prediction intervals is described in more detail in the literature, e.g., [288,289,290].
When calculating the prRI value, the number of serial measurements is crucial; the higher the n, the more certain and reliable is the value [287]. Therefore, in principle, n should be ≥5. When this is not feasible, the CVI of the analyte available from the European Federation of Clinical Chemistry and Laboratory Medicine (EFLM) Database should be used instead of the CVP value [291].
In the clinical context, RIs are used to distinguish healthy individuals, and corresponds to the specificity of the algorithm used to estimate the RIs. Specificity can be calculated from the following formula:
S p e c i f i c i t y = T N T N + F P
where TN is the number of true negatives, and FP is the number of false positives.
As shown in Figure 4A,B, the specificity of popRIs is lower than that of prRIs, indicating that the personalization of reference data for a tumor marker can enhance the specificity of its measurement, thus improving its clinical utility. A typical example is CA 19-9, commonly used for the management of colorectal and pancreatic cancer, the latter being one of the most aggressive tumors in humans. The CVI and CVG for CA 19-9 are 4.3% and 57.4%, respectively [291]. Consequently, the II for CA 19-9 is calculated as 4.3/57.4, which equals 0.075. The popRI for CA 19-9 is <37 kU/L [292]. A prRI, calculated for an individual using the CVI and an analytical CV (CVA) accepted as desirable (typically equal to half the CVI) and a hypothetical set point such as 20 kU/L obtained from 10 serial measurements, shows that the prRI range for this individual (17.7–22.3 kU/L) is significantly narrower than the popRI for CA 19-9 (0–37 kU/L) (Figure 4A). It should be noted that the range of the prRI in this case (22.3–17.7, corresponding to 4.6 kU/L) is narrower than the popRI range (37 kU/L) (Figure 4A). Due to the wide range of CVG, the popRI for CA 19-9 is broader than the prRI. Therefore, if this individual’s CA 19-9 level is 30 kU/L, it would be considered normal based on the popRI, whereas for this individual, it is actually elevated and should not be interpreted as normal. Using unsuitable RIs is analogous to following an inaccurate map and can reduce the accuracy of clinical decision-making.
The HSP may vary among individuals, reflecting the population heterogeneity. Therefore, prRIs for tumor markers reflect individual variation and differ significantly from the fixed popRI values commonly used in routine clinical practice. This example illustrates why the specificity of tumor markers is often suboptimal and why they are not commonly employed for the diagnosis of malignant conditions—the reliance on popRIs limits their diagnostic utility. However, if tumor markers were personalized, they could offer significant potential for the early diagnosis of malignancies with high specificity and sensitivity, as detailed below.

3.3.2. Personalized Decision Limits for Tumor Markers in the Diagnosis of Malignant Diseases

Although RIs are widely used in clinical practice, they are primarily intended to distinguish healthy individuals rather than to diagnose diseases. Therefore, RIs may not be suitable tools for disease diagnosis, which is instead based on decision limits (DL). The estimation of DLs is more complex than the estimation of RIs [294]. Since RIs are estimated using measurement results from healthy individuals, the estimation of DLs requires patient data, and the diagnoses of these patients must be confirmed by other techniques or parameters such as clinical findings, radiological imaging, and pathological examinations. Therefore, the selection of patients to be included in DL studies requires special attention and must be supported by well-documented clinical, radiological, pathological, and other relevant findings. Furthermore, and most importantly, while there is typically a single RI for a tumor marker, there may be multiple DLs. This makes estimating DLs more complex, requiring detailed evaluation for each tumor marker and for each particular clinical condition in which the marker is intended to be used. For example, PSA levels can be elevated in benign prostatic hyperplasia (BPH), prostate cancer, and other conditions such as prostatic inflammation or infections. However, it is well established that PSA elevations tend to be mild in BPH, whereas significantly higher levels are typically observed in prostate cancer. There is a diagnostic gray zone between a healthy prostate and one affected by malignancy, and the DLs used for diagnosing prostate cancer are often well above the upper reference limit of the RI. This concept is illustrated in Figure 5.
Although theoretically possible, the estimation of DLs for individuals is challenging in practice due to the complexity involved in defining DLs for disease diagnosis. However, this does not imply that DLs cannot be personalized. In fact, they can be individualized through an indirect approach, such as simulation studies utilizing popRIs, population-based decision limits (popDLs), and prRIs, provided that the DLs for tumor markers are well established for specific cancer types [25]. Furthermore, a direct relationship must exist between the presence of the tumor and the concentration of the corresponding tumor marker in the blood.
The difference between the upper limit of the popRI and the popDL represents the critical difference, illustrating the minimum concentration gap of tumor markers between healthy individuals and those diagnosed with cancer (Figure 5). A similar percentage change can be expected at the individual level, as formulated below [189].
p o p R C D L = L p o p R I p o p D L L p o p R I
Here, popRCDL represents the population-based relative change from the clinically significant limits of the RI to the popDLs, while LpopRI denotes the clinically significant limit of the popRI.
p r D L = L p r R I L p r R I × p o p R C D L
The specificity of an analyte pertains to its ability to correctly identify healthy individuals and is therefore associated with RIs, whereas sensitivity refers to its ability to correctly identify diseased individuals and is related to the DLs of the analyte used for disease diagnosis, as formulated below.
S e n s i t i v i t y = T P T P + F N
where TP corresponds to true positives and FN corresponds to false negatives identified by the DLs.
As shown in Figure 5, prDLs have great potential to increase the sensitivity of tumor markers and, consequently, enhance their clinical significance in the diagnosis of malignant diseases.

3.3.3. Personalized Reference Change Value for Monitoring Disease Progression and Treatment Response

RIs can be used to distinguish healthy individuals based on analyte levels; however, monitoring individuals for disease progression, evaluating treatment effectiveness, and assessing treatment side effects cannot rely solely on RIs. Therefore, a new algorithm is needed for the objective evaluation of individual monitoring in both healthy and diseased conditions. The monitoring of individuals can be performed using serial measurements of analytes during the course of disease. The significance of changes in analyte concentrations can be assessed using the reference change value (RCV), which is calculated as shown below.
R C V P = t α / 2 × 2 × C V A 2 + C V P 2
If the difference between two measurements obtained from different samples taken at the same time on different days, weeks, or months (i.e., the delta value) is lower than the RCV, then this difference can be considered insignificant at a specified probability level, such as 95% [281,295]. It indicates that this difference can be attributed to inherent variation arising from the measurement procedure and biological variation, and is therefore likely to be insignificant. Conversely, if the delta value exceeds the RCV, the difference is considered significant at a given probability level (e.g., 95%) and cannot be explained solely by analytical and/or biological variation. In such cases, the observed difference should be interpreted as resulting from other factors, such as disease progression, treatment effect, or drug-related side effects (Figure 6).

4. Conclusions

Although tumor markers are currently used primarily for monitoring malignant diseases, they possess significant potential in the broader management of cancer. More than a century of experience has shown that relying on a single biomarker is often insufficient for the early and accurate diagnosis of malignancies due to limitations in sensitivity and specificity. Therefore, combining multiple tumor markers provides a more robust diagnostic strategy. This insight calls for a new paradigm in the use of tumor markers—one that redefines their role in the early detection, monitoring, and personalized treatment of malignant diseases.
Emerging technologies such as omics platforms offer powerful avenues for the discovery of novel biomarkers for malignant diseases, while wearable biosensors enable the continuous, real-time monitoring of individuals. Given the biological heterogeneity of tumors, personalizing tumor marker interpretation becomes essential. Furthermore, artificial intelligence can integrate personal health data with wearable biosensor outputs to develop individualized algorithms for the early detection and effective management of malignancies. Together, these innovations promise to transform tumor marker applications and significantly advance personalized oncology.

Supplementary Materials

The following supporting information can be downloaded at https://www.mdpi.com/article/10.3390/biom15071011/s1. Novel Tumor Markers; Table S1. Genetic alterations; Table S2. Tissue markers; Table S3. Serum markers; Table S4. Urinary markers.

Author Contributions

Writing—original draft preparation, review and editing, visualization, I.N.S.; writing—original draft preparation, review and editing, visualization, supervision and conceptualization, A.C. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

No new data were created or analyzed in this study. Data sharing is not applicable to this article.

Acknowledgments

During the preparation of this manuscript, the authors used ChatGPT (GPT-4o) for the purpose of English language polishing.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Wild, C.P.; Weiderpass, E.; Stewart, B.W. World Cancer Report: Cancer Research for Cancer Prevention; International Agency for Research on Cancer: Lyon, France, 2020; Available online: http://publications.iarc.fr/586 (accessed on 1 May 2025).
  2. Hanna, T.P.; King, W.D.; Thibodeau, S.; Jalink, M.; Paulin, G.A.; Harvey-Jones, E.; O’Sullivan, D.E.; Booth, C.M.; Sullivan, R.; Aggarwal, A. Mortality Due to Cancer Treatment Delay: Systematic Review and Meta-Analysis. BMJ 2020, 371, m4087. [Google Scholar] [CrossRef] [PubMed]
  3. McGarvey, N.; Gitlin, M.; Fadli, E.; Chung, K.C. Increased Healthcare Costs by Later Stage Cancer Diagnosis. BMC Health Serv. Res. 2022, 22, 1155. [Google Scholar] [CrossRef] [PubMed]
  4. Costa, L.; Kumar, R.; Villarreal-Garza, C.; Sinha, S.; Saini, S.; Semwal, J.; Saxsena, V.; Zamre, V.; Chintamani, C.; Ray, M.; et al. Diagnostic Delays in Breast Cancer among Young Women: An Emphasis on Healthcare Providers. Breast 2024, 73, 103623. [Google Scholar] [CrossRef] [PubMed]
  5. Ou, X.; Chen, P.; Liu, B.-F. Liquid Biopsy on Microfluidics: From Existing Endogenous to Emerging Exogenous Biomarkers Analysis. Anal. Chem. 2025, 97, 8625–8640. [Google Scholar] [CrossRef]
  6. Sturgeon, C.M.; Duffy, M.J.; Hofmann, B.R.; Lamerz, R.; Fritsche, H.A.; Gaarenstroom, K.; Bonfrer, J.; Ecke, T.H.; Grossman, H.B.; Hayes, P.; et al. National Academy of Clinical Biochemistry Laboratory Medicine Practice Guidelines for Use of Tumor Markers in Liver, Bladder, Cervical, and Gastric Cancers. Clin. Chem. 2010, 56, e1–e48. [Google Scholar] [CrossRef]
  7. Sturgeon, C.M.; Duffy, M.J.; Stenman, U.H.; Lilja, H.; Brünner, N.; Chan, D.W.; Babaian, R.; Bast, R.C.; Dowell, B.; Esteva, F.J.; et al. National Academy of Clinical Biochemistry Laboratory Medicine Practice Guidelines for Use of Tumor Markers in Testicular, Prostate, Colorectal, Breast, and Ovarian Cancers. Clin. Chem. 2008, 54, e11–e79. [Google Scholar] [CrossRef]
  8. Meany, D.L.; Sokoll, L.J.; Chan, D.W. Early Detection of Cancer: Immunoassays for Plasma Tumor Markers. Expert. Opin. Med. Diagn. 2009, 3, 597–605. [Google Scholar] [CrossRef]
  9. Mulshine, J.L.; Scott, F. Molecular Markers in Early Cancer Detection: New Screening Tools. Chest 1995, 107, 280S–286S. [Google Scholar] [CrossRef]
  10. Molyneux, E.M.; Rochford, R.; Griffin, B.; Newton, R.; Jackson, G.; Menon, G.; Harrison, C.J.; Israels, T.; Bailey, S. Burkitt’s Lymphoma. Lancet 2012, 379, 1234–1244. [Google Scholar] [CrossRef]
  11. Shackney, S.E.; McCormack, G.W.; Cuchural, G.J. Growth Rate Patterns of Solid Tumors and Their Relation to Responsiveness to Therapy: An Analytical Review. Ann. Intern. Med. 1978, 89, 107–121. [Google Scholar] [CrossRef]
  12. Stensjøen, A.L.; Solheim, O.; Kvistad, K.A.; Håberg, A.K.; Salvesen, Ø.; Berntsen, E.M. Growth Dynamics of Untreated Glioblastomas in Vivo. Neuro Oncol. 2015, 17, 1402. [Google Scholar] [CrossRef] [PubMed]
  13. Jiang, B.; Han, D.; van der Aalst, C.M.; Lancaster, H.L.; Vonder, M.; Gratama, J.W.C.; Silva, M.; Field, J.K.; de Koning, H.J.; Heuvelmans, M.A.; et al. Lung Cancer Volume Doubling Time by Computed Tomography: A Systematic Review and Meta-Analysis. Eur. J. Cancer 2024, 212, 114339. [Google Scholar] [CrossRef]
  14. Bedia, J.S.; Jacobs, I.J.; Ryan, A.; Gentry-Maharaj, A.; Burnell, M.; Singh, N.; Manchanda, R.; Kalsi, J.K.; Dawnay, A.; Fallowfield, L.; et al. Estimating the Ovarian Cancer CA-125 Preclinical Detectable Phase, in-Vivo Tumour Doubling Time, and Window for Detection in Early Stage: An Exploratory Analysis of UKCTOCS. EBioMedicine 2025, 112, 105554. [Google Scholar] [CrossRef] [PubMed]
  15. Nathani, P.; Gopal, P.; Rich, N.; Yopp, A.; Yokoo, T.; John, B.; Marrero, J.; Parikh, N.; Singal, A.G. Hepatocellular Carcinoma Tumour Volume Doubling Time: A Systematic Review and Meta-Analysis. Gut 2021, 70, 401–407. [Google Scholar] [CrossRef]
  16. Furukawa, H.; Iwata, R.; Moriyama, N. Growth Rate of Pancreatic Adenocarcinoma: Initial Clinical Experience. Pancreas 2001, 22, 366–369. [Google Scholar] [CrossRef]
  17. Dahan, M.; Hequet, D.; Bonneau, C.; Paoletti, X.; Rouzier, R. Has Tumor Doubling Time in Breast Cancer Changed over the Past 80 Years? A Systematic Review. Cancer Med. 2021, 10, 5203–5217. [Google Scholar] [CrossRef] [PubMed]
  18. Oh, S.Y.; Lee, J.H.; Lee, H.J.; Kim, T.H.; Huh, Y.J.; Ahn, H.S.; Suh, Y.S.; Kong, S.H.; Kim, G.H.; Ahn, S.J.; et al. Natural History of Gastric Cancer: Observational Study of Gastric Cancer Patients Not Treated During Follow-Up. Ann. Surg. Oncol. 2019, 26, 2905–2911. [Google Scholar] [CrossRef]
  19. Burke, J.R.; Brown, P.; Quyn, A.; Lambie, H.; Tolan, D.; Sagar, P. Tumour Growth Rate of Carcinoma of the Colon and Rectum: Retrospective Cohort Study. BJS Open 2020, 4, 1200–1207. [Google Scholar] [CrossRef]
  20. Yeh, T.; Yeung, M.; Sherman, E.J.; Tuttle, R.M.; Sabra, M.M. Structural Doubling Time Predicts Overall Survival in Patients with Medullary Thyroid Cancer in Patients with Rapidly Progressive Metastatic Medullary Thyroid Cancer Treated with Molecular Targeted Therapies. Thyroid 2020, 30, 1112–1119. [Google Scholar] [CrossRef]
  21. Schmid, H.-P.; Mcneal, J.E.; Stamey, T.A. Observations on the Doubling Time of Prostate Cancer The Use of Serial Prostate-Specific Antigen in Patients with Untreated Disease as a Measure of Increasing Cancer Volume. Cancer 1993, 71, 2031–2040. [Google Scholar] [CrossRef]
  22. Klein, C.A. Parallel Progression of Primary Tumours and Metastases. Nat. Rev. Cancer 2009, 9, 302–312. [Google Scholar] [CrossRef] [PubMed]
  23. Oh, H.S.; Kwon, H.; Song, E.; Jeon, M.J.; Kim, T.Y.; Lee, J.H.; Kim, W.B.; Shong, Y.K.; Chung, K.W.; Baek, J.H.; et al. Tumor Volume Doubling Time in Active Surveillance of Papillary Thyroid Carcinoma. Thyroid 2019, 29, 642–649. [Google Scholar] [CrossRef] [PubMed]
  24. Catherine, S. Tumor Markers. In Tietz Fundamentals of Clinical Chemistry and Molecular Diagnostics, 9th ed.; Rifai, N., Chiu, R.W.K., Young, I., Wittwer, C.T., Eds.; Elsevier: St. Louis, MO, USA, 2024; pp. 346–371. ISBN 978-0-323-93583-8. [Google Scholar]
  25. Coskun, A.; Ertaylan, G.; Pusparum, M.; Van Hoof, R.; Kaya, Z.Z.; Khosravi, A.; Zarrabi, A. Advancing Personalized Medicine: Integrating Statistical Algorithms with Omics and Nano-Omics for Enhanced Diagnostic Accuracy and Treatment Efficacy. Biochim. Biophys. Acta Mol. Basis Dis. 2024, 1870, 167339. [Google Scholar] [CrossRef] [PubMed]
  26. 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, 1–86. [Google Scholar] [CrossRef] [PubMed]
  27. Jones, H.B. Papers ON CHEMICAL PATHOLOGY; Prefaced by the Gulstonian Lectures, Read at the Royal College of Physicians. Lancet 1847, 50, 325–330. [Google Scholar] [CrossRef]
  28. Li, M.C.; Hertz, R.; Spencer, D.B. Effect of Methotrexate Therapy upon Choriocarcinoma and Chorioadenoma. Proc. Soc. Exp. Biol. Med. 1956, 93, 361–366. [Google Scholar] [CrossRef] [PubMed]
  29. Abelev, G.I.; Perova, S.D.; Khramkova, N.I.; Postnikova, Z.A.; Irlin, I.S. Production of Embryonal Alpha-Globulin by Transplantable Mouse Hepatomas. Transplantation 1963, 1, 174–180. [Google Scholar] [CrossRef]
  30. Gold, P.; Freedman, S.O. Demonstration of Tumor-specific Antigens in Human Colonic Carcinomata by Immunological Tolerance and Absorption Techniques. J. Exp. Med. 1965, 121, 439–462. [Google Scholar] [CrossRef]
  31. Moore, B.W.; McGregor, D. Chromatographic and Electrophoretic Fractionation of Soluble Proteins of Brain and Liver. J. Biol. Chem. 1965, 240, 1647–1653. [Google Scholar] [CrossRef]
  32. Blaschko, H.; Comline, R.S.; Schneider, F.H.; Silver, M.; Smith, A.D. Secretion of a Chromaffin Granule Protein, Chromogranin, from the Adrenal Gland after Splanchnic Stimulation. Nature 1967, 215, 58–59. [Google Scholar] [CrossRef]
  33. Tashjian, A.H., Jr.; Melvin, K.E. Medullary Carcinoma of the Thyroid Gland: Studies of Thyrocalcitonin in Plasma and Tumor Extracts. N. Engl. J. Med. 1968, 279, 279–283. [Google Scholar] [CrossRef] [PubMed]
  34. Van Herle, A.J.; Uller, R.P. Elevated Serum Thyroglobulin. A Marker of Metastases in Differentiated Thyroid Carcinomas. J. Clin. Investig. 1975, 56, 272–277. [Google Scholar] [CrossRef]
  35. Kato, H.; Torigoe, T. Radioimmunoassay for tumor antigen of human cervical squamous cell carcinoma. Cancer 1977, 40, 1621–1628. [Google Scholar] [CrossRef]
  36. Wang, M.C.; Valenzuela, L.A.; Murphy, G.P.; Chu, T.M. Purification of a Human Prostate Specific Antigen. Investig. Urol. 1979, 17, 159–163. [Google Scholar] [PubMed]
  37. Koprowski, H.; Steplewski, Z.; Mitchell, K.; Herlyn, M.; Herlyn, D.; Fuhrer, P. Colorectal Carcinoma Antigens Detected by Hybridoma Antibodies. Somat. Cell Genet. 1979, 5, 957–971. [Google Scholar] [CrossRef]
  38. Bast, R.C.; Feeney, M.; Lazarus, H.; Nadler, L.M.; Colvin, R.B.; Knapp, R.C. Reactivity of a Monoclonal Antibody with Human Ovarian Carcinoma. J. Clin. Investig. 1981, 68, 1331–1337. [Google Scholar] [CrossRef]
  39. Kufe, D.; Inghirami, G.; Abe, M.; Hayes, D.; Justi-Wheeler, H.; Schlom, J. Differential Reactivity of a Novel Monoclonal Antibody (DF3) with Human Malignant versus Benign Breast Tumors. Hybridoma 1984, 3, 223–232. [Google Scholar] [CrossRef] [PubMed]
  40. Lappöhn, R.E.; Burger, H.G.; Bouma, J.; Bangah, M.; Krans, M.; De Bruijn, H.W.A. Inhibin as a Marker for Granulosa-Cell Tumors. N. Engl. J. Med. 1989, 321, 790–793. [Google Scholar] [CrossRef]
  41. Kirchhoff, C.; Habben, I.; Iveli, R.; Krull, N. A Major Human Epididymis-Specific CDNA Encodes a Protein with Sequence Homology to Extracellular Proteinase Inhibitors. Biol. Reprod. 1991, 45, 350–357. [Google Scholar] [CrossRef]
  42. Pujol, J.L.; Grenier, J.; Daurès, J.P.; Daver, A.; Pujol, H.; Michel, F.B. Serum fragment of cytokeratin subunit 19 measured by CYFRA 21-1 immunoradiometric assay as a marker of lung cancer. Cancer Res. 1993, 53, 61–66. [Google Scholar] [PubMed]
  43. Liu, B.; Zhou, H.; Tan, L.; Siu, K.T.H.; Guan, X.Y. Exploring Treatment Options in Cancer: Tumor Treatment Strategies. Signal Transduct. Target. Ther. 2024, 9, 1–44. [Google Scholar] [CrossRef] [PubMed]
  44. Bai, R.; Meng, Y.; Cui, J. Therapeutic Strategies Targeting Metabolic Characteristics of Cancer Cells. Crit. Rev. Oncol. Hematol. 2023, 187, 104037. [Google Scholar] [CrossRef] [PubMed]
  45. Kreuzaler, P.; Panina, Y.; Segal, J.; Yuneva, M. Adapt and Conquer: Metabolic Flexibility in Cancer Growth, Invasion and Evasion. Mol. Metab. 2020, 33, 83–101. [Google Scholar] [CrossRef] [PubMed]
  46. Tufail, M.; Jiang, C.-H.; Li, N. Altered Metabolism in Cancer: Insights into Energy Pathways and Therapeutic Targets. Mol. Cancer 2024, 23, 1–40. [Google Scholar] [CrossRef]
  47. Reznik, E.; Luna, A.; Aksoy, B.A.; Liu, E.M.; La, K.; Ostrovnaya, I.; Creighton, C.J.; Hakimi, A.A.; Sander, C. A Landscape of Metabolic Variation across Tumor Types. Cell Syst. 2018, 6, 301–313. [Google Scholar] [CrossRef]
  48. Ramírez-Carmona, W.; Díaz-Fabregat, B.; Yuri Yoshigae, A.; Musa de Aquino, A.; Scarano, W.R.; de Souza Castilho, A.C.; Avansini Marsicano, J.; Leal do Prado, R.; Pessan, J.P.; de Oliveira Mendes, L. Are Serum Ferritin Levels a Reliable Cancer Biomarker? A Systematic Review and Meta-Analysis. Nutr. Cancer 2022, 74, 1917–1926. [Google Scholar] [CrossRef]
  49. Konety, B.R.; Nguyen, T.S.T.; Brenes, G.; Sholder, A.; Lewis, N.; Bastacky, S.; Getzenberg, R.H. Clinical usefulness of the novel marker BLCA-4 for the detection of bladder cancer. J. Urol. 2000, 164, 634–639. [Google Scholar] [CrossRef]
  50. Fontana, V.; Pistillo, M.P.; Vigani, A.; Canessa, P.A.; Berisso, G.; Giannoni, U.; Roncella, S. Potential role of serum mesothelin in predicting survival of patients with malignant pleural mesothelioma. Oncol. Lett. 2021, 21, 128. [Google Scholar] [CrossRef]
  51. Cheng, Y.; Wang, C.; Wang, Y.; Dai, L. Soluble PD-L1 as a Predictive Biomarker in Lung Cancer: A Systematic Review and Meta-Analysis. Future Oncol. 2022, 18, 261–273. [Google Scholar] [CrossRef]
  52. Tsé, C.; Gauchez, A.S.; Jacot, W.; Lamy, P.J. HER2 shedding and serum HER2 extracellular domain: Biology and clinical utility in breast cancer. Cancer Treat. Rev. 2012, 38, 133–142. [Google Scholar] [CrossRef]
  53. Perrier, A.; Gligorov, J.; Lefèvre, G.; Boissan, M. The Extracellular Domain of Her2 in Serum as a Biomarker of Breast Cancer. Lab. Investig. 2018, 98, 696–707. [Google Scholar] [CrossRef] [PubMed]
  54. Duffy, M.J. Clinical Uses of Tumor Markers: A Critical Review. Crit. Rev. Clin. Lab. Sci. 2001, 38, 225–262. [Google Scholar] [CrossRef] [PubMed]
  55. Chan, D.W.; Booth, R.A.; Diamandis, E.P. Tumor Markers. In Tietz Textbook of Clinical Chemistry and Molecular Diagnostics, 4th ed.; Burtis, C.A., Ashwood, E.R., Bruns, D.E., Eds.; Elsevier Saunders: St. Louis, MO, USA, 2006; pp. 745–795. ISBN 0-7216-0189-8. [Google Scholar]
  56. Głowska-Ciemny, J.; Szmyt, K.; Kuszerska, A.; Rzepka, R.; von Kaisenberg, C.; Kocyłowski, R. Fetal and Placental Causes of Elevated Serum Alpha-Fetoprotein Levels in Pregnant Women. J. Clin. Med. 2024, 13, 466. [Google Scholar] [CrossRef] [PubMed]
  57. Tzartzeva, K.; Obi, J.; Rich, N.E.; Parikh, N.D.; Marrero, J.A.; Yopp, A.; Waljee, A.K.; Singal, A.G. Surveillance Imaging and Alpha Fetoprotein for Early Detection of Hepatocellular Carcinoma in Patients With Cirrhosis: A Meta-Analysis. Gastroenterology 2018, 154, 1706–1718. [Google Scholar] [CrossRef]
  58. Debes, J.D.; Romagnoli, P.A.; Prieto, J.; Arrese, M.; Mattos, A.Z.; Boonstra, A. Serum Biomarkers for the Prediction of Hepatocellular Carcinoma. Cancers 2021, 13, 1681. [Google Scholar] [CrossRef]
  59. Kudo, M. Urgent Global Need for PIVKA-II and AFP-L3 Measurements for Surveillance and Management of Hepatocellular Carcinoma. Liver Cancer 2024, 13, 113–118. [Google Scholar] [CrossRef]
  60. Liu, Y.; Jiang, W.; Li, X.; Zhao, H.; Wang, S. The Diagnostic Performance of AFP, AFP-L3, DCP, CA199, and Their Combination for Primary Liver Cancer. J. Hepatocell. Carcinoma 2025, 12, 513–526. [Google Scholar] [CrossRef]
  61. Jansen, K.; Kornfeld, L.; Lennartz, M.; Dwertmann Rico, S.; Kind, S.; Reiswich, V.; Viehweger, F.; Bawahab, A.A.; Fraune, C.; Gorbokon, N.; et al. Carcinoembryonic Antigen Expression in Human Tumors: A Tissue Microarray Study on 13,725 Tumors. Cancers 2024, 16, 4052. [Google Scholar] [CrossRef]
  62. Zhang, C.; Zheng, W.; Lv, Y.; Shan, L.; Xu, D.; Pan, Y.; Zhu, H.; Qi, H. Postoperative Carcinoembryonic Antigen (CEA) Levels Predict Outcomes after Resection of Colorectal Cancer in Patients with Normal Preoperative CEA Levels. Transl. Cancer Res. 2020, 9, 111–118. [Google Scholar] [CrossRef]
  63. Hull, M.A.; Rees, C.J.; Sharp, L.; Koo, S. A Risk-Stratified Approach to Colorectal Cancer Prevention and Diagnosis. Nat. Rev. Gastroenterol. Hepatol. 2020, 17, 773–780. [Google Scholar] [CrossRef]
  64. Guo, J.; Yu, J.; Song, X.; Mi, H. Serum CA125, CA199 and CEA Combined Detection for Epithelial Ovarian Cancer Diagnosis: A Meta-Analysis. Open Med. 2017, 12, 1. [Google Scholar] [CrossRef]
  65. Wang, H.; Jin, W.; Wan, C.; Zhu, C. Diagnostic Value of Combined Detection of CA72-4, CA19-9, and Carcinoembryonic Antigen Comparing to CA72-4 Alone in Gastric Cancer: A Systematic Review and Meta-Analysis. Transl. Cancer Res. 2022, 11, 848–856. [Google Scholar] [CrossRef]
  66. Zhan, C.H.; Liu, G.J. Diagnostic Value of a Combined Serum &alpha;-Hydroxybutyrate Dehydrogenase, Carcinoembryonic Antigen and Glycoantigen 125 Test for Early-Stage Breast Cancer. Breast Cancer Targets Ther. 2023, 15, 617–623. [Google Scholar] [CrossRef]
  67. Izawa, A.; Hara, Y.; Horita, N.; Muraoka, S.; Kaneko, M.; Kaneko, A.; Somekawa, K.; Hirata, M.; Otsu, Y.; Matsumoto, H.; et al. Improved Diagnostic Accuracy with Three Lung Tumor Markers Compared to Six-Marker Panel. Transl. Lung Cancer Res. 2024, 13, 503–511. [Google Scholar] [CrossRef] [PubMed]
  68. Zhou, C.M.; Zhao, S.H. Evaluation of the Value of Combined Detection of Tumor Markers CA724, Carcinoembryonic Antigen, CA242, and CA19-9 in Gastric Cancer. World J. Gastrointest. Oncol. 2024, 16, 1737–1744. [Google Scholar] [CrossRef] [PubMed]
  69. Li, L.; Xu, Y.; Wang, Y.; Zhang, Q.; Wang, Y.; Xu, C. The Diagnostic and Prognostic Value of the Combination of Tumor M2-Pyruvate Kinase, Carcinoembryonic Antigen, and Cytokeratin 19 Fragment in Non-Small Cell Lung Cancer. Technol. Cancer Res. Treat. 2024, 23, 15330338241265983. [Google Scholar] [CrossRef] [PubMed]
  70. Molina, R.; Marrades, R.M.; Augé, J.M.; Escudero, J.M.; Viñolas, N.; Reguart, N.; Ramirez, J.; Filella, X.; Molins, L.; Agustí, A. Assessment of a Combined Panel of Six Serum Tumor Markers for Lung Cancer. Am. J. Respir. Crit. Care Med. 2016, 193, 427–437. [Google Scholar] [CrossRef]
  71. Nagpal, M.; Singh, S.; Singh, P.; Chauhan, P.; Zaidi, M. Tumor Markers: A Diagnostic Tool. Natl. J. Maxillofac. Surg. 2016, 7, 17. [Google Scholar] [CrossRef]
  72. Babkina, A.S.; Lyubomudrov, M.A.; Golubev, M.A.; Pisarev, M.V.; Golubev, A.M. Neuron-Specific Enolase—What Are We Measuring? Int. J. Mol. Sci. 2024, 25, 5040. [Google Scholar] [CrossRef]
  73. Wang, W.; Yang, C.; Wang, T.; Deng, H. Complex Roles of Nicotinamide N-Methyltransferase in Cancer Progression. Cell Death Dis. 2022, 13, 267. [Google Scholar] [CrossRef]
  74. Jiang, T.; Zeng, Q.; He, J. Do Alkaline Phosphatases Have Great Potential in the Diagnosis, Prognosis, and Treatment of Tumors? Transl. Cancer Res. 2023, 12, 2932–2945. [Google Scholar] [CrossRef] [PubMed]
  75. Ma, H.; Hei, A.; Zhou, J.; He, E.; Skog, S.; Li, J. Serum Thymidine Kinase 1 Protein Concentration for Predicting Early Progression and Monitoring the Response to TACE in Hepatocellular Carcinomas: A Network Meta-Analysis. Future Sci. OA 2021, 7, FSO717. [Google Scholar] [CrossRef]
  76. Lilja, H.; Ulmert, D.; Vickers, A.J. Prostate-Specific Antigen and Prostate Cancer: Prediction, Detection and Monitoring. Nat. Rev. Cancer 2008, 8, 268–278. [Google Scholar] [CrossRef] [PubMed]
  77. Garrido, M.M.; Bernardino, R.M.; Marta, J.C.; Holdenrieder, S.; Guimarães, J.T. Tumour Markers in Prostate Cancer: The Post-Prostate-Specific Antigen Era. Ann. Clin. Biochem. 2022, 59, 46–58. [Google Scholar] [CrossRef]
  78. Benson, M.C.; Whang, I.S.; Pantuck, A.; Ring, K.; Kaplan, S.A.; Olsson, C.A.; Cooner, W.H. Prostate Specific Antigen Density: A Means of Distinguishing Benign Prostatic Hypertrophy and Prostate Cancer. J. Urol. 1992, 147, 815–816. [Google Scholar] [CrossRef]
  79. Kachuri, L.; Hoffmann, T.J.; Jiang, Y.; Berndt, S.I.; Shelley, J.P.; Schaffer, K.R.; Machiela, M.J.; Freedman, N.D.; Huang, W.Y.; Li, S.A.; et al. Genetically Adjusted PSA Levels for Prostate Cancer Screening. Nat. Med. 2023, 29, 1412–1423. [Google Scholar] [CrossRef] [PubMed]
  80. Charing Cross Gestational Trophoblast Disease Service. Available online: https://www.hmole-chorio.org.uk/ (accessed on 7 April 2025).
  81. Pelizzo, M.R.; Mazza, E.I.; Mian, C.; Merante Boschin, I. Medullary Thyroid Carcinoma. Expert. Rev. Anticancer Ther. 2023, 23, 943–957. [Google Scholar] [CrossRef]
  82. Censi, S.; Manso, J.; Mian, C. Other Markers of Medullary Thyroid Cancer, Not Only Calcitonin. Eur. J. Endocrinol. 2023, 188, R1–R13. [Google Scholar] [CrossRef]
  83. Ni, J.; Tu, P.; Ling, Y. Gender and Tumor Size-Specific Calcitonin Cutoff Value for Diagnosing MTC in 10,618 Patients with Thyroid Nodule Surgery. Endocrine 2024, 86, 1097–1109. [Google Scholar] [CrossRef]
  84. Giovanella, L.; Garo, M.L.; Ceriani, L.; Paone, G.; Campenni’, A.; D’Aurizio, F. Procalcitonin as an Alternative Tumor Marker of Medullary Thyroid Carcinoma. J. Clin. Endocrinol. Metab. 2021, 106, 3634–3643. [Google Scholar] [CrossRef]
  85. Luo, G.; Guo, M.; Jin, K.; Liu, Z.; Liu, C.; Cheng, H.; Lu, Y.; Long, J.; Liu, L.; Xu, J.; et al. Optimize CA19-9 in Detecting Pancreatic Cancer by Lewis and Secretor Genotyping. Pancreatology 2016, 16, 1057–1062. [Google Scholar] [CrossRef] [PubMed]
  86. Luo, G.; Jin, K.; Deng, S.; Cheng, H.; Fan, Z.; Gong, Y.; Qian, Y.; Huang, Q.; Ni, Q.; Liu, C.; et al. Roles of CA19-9 in Pancreatic Cancer: Biomarker, Predictor and Promoter. Biochim. Biophys. Acta Rev. Cancer 2021, 1875, 188409. [Google Scholar] [CrossRef]
  87. Yang, J.; Xu, R.; Wang, C.; Qiu, J.; Ren, B.; You, L. Early Screening and Diagnosis Strategies of Pancreatic Cancer: A Comprehensive Review. Cancer Commun. 2021, 41, 1257–1274. [Google Scholar] [CrossRef]
  88. Kaur, S.; Smith, L.M.; Patel, A.; Menning, M.; Watley, D.C.; Malik, S.S.; Krishn, S.R.; Mallya, K.; Aithal, A.; Sasson, A.R.; et al. A Combination of MUC5AC and CA19-9 Improves the Diagnosis of Pancreatic Cancer: A Multicenter Study. Am. J. Gastroenterol. 2017, 112, 172–183. [Google Scholar] [CrossRef] [PubMed]
  89. Zhang, X.Y.; Hong, L.L.; Ling, Z.Q. MUC16/CA125 in Cancer: New Advances. Clin. Chim. Acta 2025, 565, 119981. [Google Scholar] [CrossRef] [PubMed]
  90. Ghose, A.; McCann, L.; Makker, S.; Mukherjee, U.; Gullapalli, S.V.N.; Erekkath, J.; Shih, S.; Mahajan, I.; Sanchez, E.; Uccello, M.; et al. Diagnostic Biomarkers in Ovarian Cancer: Advances beyond CA125 and HE4. Ther. Adv. Med. Oncol. 2024, 16, 17588359241233225. [Google Scholar] [CrossRef]
  91. Mukama, T.; Fortner, R.T.; Katzke, V.; Hynes, L.C.; Petrera, A.; Hauck, S.M.; Johnson, T.; Schulze, M.; Schiborn, C.; Rostgaard-Hansen, A.L.; et al. Prospective Evaluation of 92 Serum Protein Biomarkers for Early Detection of Ovarian Cancer. Br. J. Cancer 2022, 126, 1301–1309. [Google Scholar] [CrossRef]
  92. Nath, S.; Mukherjee, P. MUC1: A Multifaceted Oncoprotein with a Key Role in Cancer Progression. Trends Mol. Med. 2014, 20, 332–342. [Google Scholar] [CrossRef]
  93. Duffy, M.J.; Evoy, D.; McDermott, E.W. CA 15-3: Uses and Limitation as a Biomarker for Breast Cancer. Clin. Chim. Acta 2010, 411, 1869–1874. [Google Scholar] [CrossRef]
  94. Zhao, W.; Li, X.; Wang, W.; Chen, B.; Wang, L.; Zhang, N.; Wang, Z.; Yang, Q. Association of Preoperative Serum Levels of CEA and CA15-3 with Molecular Subtypes of Breast Cancer. Dis. Markers 2021, 2021, 5529106. [Google Scholar] [CrossRef]
  95. Ryu, J.M.; Kang, D.; Cho, J.; Lee, J.E.; Kim, S.W.; Nam, S.J.; Lee, S.K.; Kim, Y.J.; Im, Y.H.; Ahn, J.S.; et al. Prognostic Impact of Elevation of Cancer Antigen 15-3 (CA15-3) in Patients With Early Breast Cancer With Normal Serum CA15-3 Level. J. Breast Cancer 2023, 26, 126. [Google Scholar] [CrossRef]
  96. Sekacheva, M.; Boroda, A.; Fatyanova, A.; Rozhkov, A.; Bagmet, N. Clinical Validation of the Novel CLIA-CA-62 Assay Efficacy for Early-Stage Breast Cancer Detection. Front. Oncol. 2023, 13, 1009863. [Google Scholar] [CrossRef]
  97. Tcherkassova, J.; Prostyakova, A.; Tsurkan, S.; Ragoulin, V.; Boroda, A.; Sekacheva, M. Diagnostic Efficacy of the New Prospective Biomarker’s Combination CA 15-3 and CA-62 for Early-Stage Breast Cancer Detection: Results of the Blind Prospective-Retrospective Clinical Study. Cancer Biomark. 2022, 35, 57–69. [Google Scholar] [CrossRef] [PubMed]
  98. Masuda, T.; Hayashi, N.; Iguchi, T.; Ito, S.; Eguchi, H.; Mimori, K. Clinical and Biological Significance of Circulating Tumor Cells in Cancer. Mol. Oncol. 2016, 10, 408–417. [Google Scholar] [CrossRef]
  99. Xu, W.; Yuan, F. Detection of Circulating Tumor Cells in the Prognostic Significance of Patients With Breast Cancer: A Retrospective Study. J. Clin. Lab. Anal. 2025, 39, e25126. [Google Scholar] [CrossRef] [PubMed]
  100. Bae, S.Y.; Kamalanathan, K.J.; Galeano-Garces, C.; Konety, B.R.; Antonarakis, E.S.; Parthasarathy, J.; Hong, J.; Drake, J.M. Dissemination of Circulating Tumor Cells in Breast and Prostate Cancer: Implications for Early Detection. Endocrinology 2024, 165, bqae022. [Google Scholar] [CrossRef] [PubMed]
  101. Gu, X.; Wei, S.; Lv, X. Circulating Tumor Cells: From New Biological Insights to Clinical Practice. Signal Transduct. Target. Ther. 2024, 9, 1–35. [Google Scholar] [CrossRef]
  102. Goldkorn, A.; Tangen, C.; Plets, M.; Bsteh, D.; Xu, T.; Pinski, J.K.; Ingles, S.; Triche, T.J.; MacVicar, G.R.; Vaena, D.A.; et al. Circulating Tumor Cell Count and Overall Survival in Patients With Metastatic Hormone-Sensitive Prostate Cancer. JAMA Netw. Open 2024, 7, e2437871. [Google Scholar] [CrossRef]
  103. Choi, S.-W.; Sun, A.K.; Cheung, J.P.-Y.; Ho, J.C.-Y. Circulating Tumour Cells in the Prediction of Bone Metastasis. Cancers 2024, 16, 252. [Google Scholar] [CrossRef]
  104. Magro, D.; Venezia, M.; Rita Balistreri, C. The Omics Technologies and Liquid Biopsies: Advantages, Limitations, Applications. Med. Omics 2024, 11, 100039. [Google Scholar] [CrossRef]
  105. Krebs, M.G.; Sloane, R.; Priest, L.; Lancashire, L.; Hou, J.M.; Greystoke, A.; Ward, T.H.; Ferraldeschi, R.; Hughes, A.; Clack, G.; et al. Evaluation and Prognostic Significance of Circulating Tumor Cells in Patients with Non-Small-Cell Lung Cancer. J. Clin. Oncol. 2011, 29, 1556–1563. [Google Scholar] [CrossRef] [PubMed]
  106. Fiorelli, A.; Accardo, M.; Carelli, E.; Angioletti, D.; Santini, M.; Di Domenico, M. Circulating Tumor Cells in Diagnosing Lung Cancer: Clinical and Morphologic Analysis. Ann. Thorac. Surg. 2015, 99, 1899–1905. [Google Scholar] [CrossRef] [PubMed]
  107. Cabel, L.; Proudhon, C.; Gortais, H.; Loirat, D.; Coussy, F.; Pierga, J.Y.; Bidard, F.C. Circulating Tumor Cells: Clinical Validity and Utility. Int. J. Clin. Oncol. 2017, 22, 421–430. [Google Scholar] [CrossRef]
  108. Sewpersad, S.; Pillay, T.S. Historical Perspectives in Clinical Pathology: Bence Jones Protein—Early Urine Chemistry and the Impact on Modern Day Diagnostics. J. Clin. Pathol. 2021, 74, 212–215. [Google Scholar] [CrossRef]
  109. Mizuno, T.; Goto, T.; Shimojo, K.; Watanabe, N.; Tanaka, T. Clinical Utility of Tumor Markers. Open J. Pathol. 2021, 11, 38–57. [Google Scholar] [CrossRef]
  110. Olivier, M.; Asmis, R.; Hawkins, G.A.; Howard, T.D.; Cox, L.A. The Need for Multi-Omics Biomarker Signatures in Precision Medicine. Int. J. Mol. Sci. 2019, 20, 4781. [Google Scholar] [CrossRef]
  111. Dhillon, A.; Singh, A.; Bhalla, V.K. A Systematic Review on Biomarker Identification for Cancer Diagnosis and Prognosis in Multi-Omics: From Computational Needs to Machine Learning and Deep Learning. Arch. Comput. Methods Eng. 2022, 30, 917–949. [Google Scholar] [CrossRef]
  112. Agarwala, P.K.; Aneja, R.; Kapoor, S. Lipidomic Landscape in Cancer: Actionable Insights for Membrane-Based Therapy and Diagnoses. Med. Res. Rev. 2022, 42, 983–1018. [Google Scholar] [CrossRef]
  113. Quezada, H.; Guzmán-Ortiz, A.L.; Díaz-Sánchez, H.; Valle-Rios, R.; Aguirre-Hernández, J. Omics-Based Biomarkers: Current Status and Potential Use in the Clinic. Bol. Med. Hosp. Infant. Mex. 2017, 74, 219–226. [Google Scholar] [CrossRef]
  114. Rossi, C.; Cicalini, I.; Cufaro, M.C.; Consalvo, A.; Upadhyaya, P.; Sala, G.; Antonucci, I.; Del Boccio, P.; Stuppia, L.; De Laurenzi, V. Breast Cancer in the Era of Integrating “Omics” Approaches. Oncogenesis 2022, 11, 1–13. [Google Scholar] [CrossRef]
  115. Thomas, N.E.; Edmiston, S.N.; Tsai, Y.S.; Parker, J.S.; Googe, P.B.; Busam, K.J.; Scott, G.A.; Zedek, D.C.; Parrish, E.A.; Hao, H.; et al. Utility of TERT Promoter Mutations for Cutaneous Primary Melanoma Diagnosis. Am. J. Dermatopathol. 2019, 41, 264–272. [Google Scholar] [CrossRef] [PubMed]
  116. Jin, A.; Xu, J.; Wang, Y. The Role of TERT Promoter Mutations in Postoperative and Preoperative Diagnosis and Prognosis in Thyroid Cancer. Medicine 2018, 97, e11548. [Google Scholar] [CrossRef] [PubMed]
  117. Muralidharan, K.; Yekula, A.; Small, J.L.; Rosh, Z.S.; Kang, K.M.; Wang, L.; Lau, S.; Zhang, H.; Lee, H.; Bettegowda, C.; et al. TERT Promoter Mutation Analysis for Blood-Based Diagnosis and Monitoring of Gliomas. Clin. Cancer Res. 2021, 27, 169–178. [Google Scholar] [CrossRef]
  118. Cheng, L.; Zhang, S.; Wang, M.; Lopez-Beltran, A. Biological and Clinical Perspectives of TERT Promoter Mutation Detection on Bladder Cancer Diagnosis and Management. Hum. Pathol. 2023, 133, 56–75. [Google Scholar] [CrossRef] [PubMed]
  119. Ruan, X.; Ye, Y.; Cheng, W.; Xu, L.; Huang, M.; Chen, Y.; Zhu, J.; Lu, X.; Yan, F. Multi-Omics Integrative Analysis of Lung Adenocarcinoma: An in Silico Profiling for Precise Medicine. Front. Med. 2022, 9, 894338. [Google Scholar] [CrossRef]
  120. Yao, M.; Fu, L.; Liu, X.; Zheng, D. In-Silico Multi-Omics Analysis of the Functional Significance of Calmodulin 1 in Multiple Cancers. Front. Genet. 2022, 12, 793508. [Google Scholar] [CrossRef] [PubMed]
  121. Zalfa, F.; Perrone, M.G.; Ferorelli, S.; Laera, L.; Pierri, C.L.; Tolomeo, A.; Dimiccoli, V.; Perrone, G.; De Grassi, A.; Scilimati, A. Genome-Wide Identification and Validation of Gene Expression Biomarkers in the Diagnosis of Ovarian Serous Cystadenocarcinoma. Cancers 2022, 14, 3764. [Google Scholar] [CrossRef]
  122. Christou, C.; Stylianou, A.; Gkretsi, V. Midkine (MDK) in Hepatocellular Carcinoma: More than a Biomarker. Cells 2024, 13, 136. [Google Scholar] [CrossRef]
  123. Christensen, M.V.; Høgdall, C.K.; Umsen, K.M.J.; Høgdall, E.V.S. Annexin A2 and Cancer: A Systematic Review. Int. J. Oncol. 2018, 52, 5–18. [Google Scholar] [CrossRef]
  124. Capone, E.; Iacobelli, S.; Sala, G. Role of Galectin 3 Binding Protein in Cancer Progression: A Potential Novel Therapeutic Target. J. Transl. Med. 2021, 19, 1–18. [Google Scholar] [CrossRef]
  125. Makarem, A. El Diagnostic Significance of Plasma Osteopontin in Hepatitis C Virus-Related Hepatocellular Carcinoma. Ann. Hepatol. 2011, 10, 296–305. [Google Scholar] [CrossRef] [PubMed]
  126. Zhu, M.; Zheng, J.; Wu, F.; Kang, B.; Liang, J.; Heskia, F.; Zhang, X.; Shan, Y. OPN Is a Promising Serological Biomarker for Hepatocellular Carcinoma Diagnosis. J. Med. Virol. 2020, 92, 3596–3603. [Google Scholar] [CrossRef]
  127. Popovics, P.; Awadallah, W.N.; Kohrt, S.E.; Case, T.C.; Miller, N.L.; Ricke, E.A.; Huang, W.; Ramirez-Solano, M.; Liu, Q.; Vezina, C.M.; et al. Prostatic Osteopontin Expression Is Associated with Symptomatic Benign Prostatic Hyperplasia. Prostate 2020, 80, 731–741. [Google Scholar] [CrossRef]
  128. Fouad, S.A.; Mohamed, N.A.G.; Fawzy, M.W.; Moustafa, D.A. Plasma Osteopontin Level in Chronic Liver Disease and Hepatocellular Carcinoma. Hepat. Mon. 2015, 15, e30753. [Google Scholar] [CrossRef] [PubMed]
  129. Ghazanfar, S.; Fatima, I.; Aslam, M.; Musharraf, S.G.; Sherman, N.E.; Moskaluk, C.; Fox, J.W.; Akhtar, M.W.; Sadaf, S. Identification of Actin Beta-like 2 (ACTBL2) as Novel, Upregulated Protein in Colorectal Cancer. J. Proteom. 2017, 152, 33–40. [Google Scholar] [CrossRef]
  130. Lobo, M.D.P.; Moreno, F.B.M.B.; Souza, G.H.M.F.; Verde, S.M.M.L.; Moreira, R.d.A.; Monteiro-Moreira, A.C.d.O. Label-Free Proteome Analysis of Plasma from Patients with Breast Cancer: Stage-Specific Protein Expression. Front. Oncol. 2017, 7, 218323. [Google Scholar] [CrossRef] [PubMed]
  131. Liu, W.; Li, J.; Zhang, P.; Hou, Q.; Feng, S.; Liu, L.; Cui, D.; Shi, H.; Fu, Y.; Luo, Y. A Novel Pan-Cancer Biomarker Plasma Heat Shock Protein 90alpha and Its Diagnosis Determinants in Clinic. Cancer Sci. 2019, 110, 2941–2959. [Google Scholar] [CrossRef] [PubMed]
  132. Shen, Q.; Fan, J.; Yang, X.R.; Tan, Y.; Zhao, W.; Xu, Y.; Wang, N.; Niu, Y.; Wu, Z.; Zhou, J.; et al. Serum DKK1 as a Protein Biomarker for the Diagnosis of Hepatocellular Carcinoma: A Large-Scale, Multicentre Study. Lancet Oncol. 2012, 13, 817–826. [Google Scholar] [CrossRef]
  133. Guo, X.; Lv, X.; Fang, C.; Lv, X.; Wang, F.; Wang, D.; Zhao, J.; Ma, Y.; Xue, Y.; Bai, Q.; et al. Dysbindin as a Novel Biomarker for Pancreatic Ductal Adenocarcinoma Identified by Proteomic Profiling. Int. J. Cancer 2016, 139, 1821–1829. [Google Scholar] [CrossRef]
  134. Zhang, J.; Wang, Y.; Zhao, T.; Li, Y.; Tian, L.; Zhao, J.; Zhang, J. Evaluation of Serum MUC5AC in Combination with CA19-9 for the Diagnosis of Pancreatic Cancer. World J. Surg. Oncol. 2020, 18, 1–7. [Google Scholar] [CrossRef]
  135. Capello, M.; Bantis, L.E.; Scelo, G.; Zhao, Y.; Li, P.; Dhillon, D.S.; Patel, N.J.; Kundnani, D.L.; Wang, H.; Abbruzzese, J.L.; et al. Sequential Validation of Blood-Based Protein Biomarker Candidates for Early-Stage Pancreatic Cancer. J. Natl. Cancer Inst. 2017, 109, djw266. [Google Scholar] [CrossRef] [PubMed]
  136. Ortega-Martínez, I.; Gardeazabal, J.; Erramuzpe, A.; Sanchez-Diez, A.; Cortés, J.; García-Vázquez, M.D.; Pérez-Yarza, G.; Izu, R.; Luís Díaz-Ramón, J.; de la Fuente, I.M.; et al. Vitronectin and Dermcidin Serum Levels Predict the Metastatic Progression of AJCC I–II Early-Stage Melanoma. Int. J. Cancer 2016, 139, 1598–1607. [Google Scholar] [CrossRef]
  137. Greco, M.; Mitri, M.D.; Chiriacò, F.; Leo, G.; Brienza, E.; Maffia, M. Serum Proteomic Profile of Cutaneous Malignant Melanoma and Relation to Cancer Progression: Association to Tumor Derived Alpha-N-Acetylgalactosaminidase Activity. Cancer Lett. 2009, 283, 222–229. [Google Scholar] [CrossRef] [PubMed]
  138. Gao, L.; Lin, Y.; Yue, P.; Li, S.; Zhang, Y.; Mi, N.; Bai, M.; Fu, W.; Xia, Z.; Jiang, N.; et al. Identification of a Novel Bile Marker Clusterin and a Public Online Prediction Platform Based on Deep Learning for Cholangiocarcinoma. BMC Med. 2023, 21, 1–15. [Google Scholar] [CrossRef]
  139. Sreekumar, A.; Poisson, L.M.; Rajendiran, T.M.; Khan, A.P.; Cao, Q.; Yu, J.; Laxman, B.; Mehra, R.; Lonigro, R.J.; Li, Y.; et al. Metabolomic Profiles Delineate Potential Role for Sarcosine in Prostate Cancer Progression. Nature 2009, 457, 910–914. [Google Scholar] [CrossRef]
  140. Khan, A.P.; Rajendiran, T.M.; Ateeq, B.; Asangani, I.A.; Athanikar, J.N.; Yocum, A.K.; Mehra, R.; Siddiqui, J.; Palapattu, G.; Wei, J.T.; et al. The Role of Sarcosine Metabolism in Prostate Cancer Progression. Neoplasia 2013, 15, 491-IN13. [Google Scholar] [CrossRef] [PubMed]
  141. Cernei, N.; Heger, Z.; Gumulec, J.; Zitka, O.; Masarik, M.; Babula, P.; Eckschlager, T.; Stiborova, M.; Kizek, R.; Adam, V. Sarcosine as a Potential Prostate Cancer Biomarker—A Review. Int. J. Mol. Sci. 2013, 14, 13893–13908. [Google Scholar] [CrossRef]
  142. Du, X.; Hu, H. The Roles of 2-Hydroxyglutarate. Front. Cell Dev. Biol. 2021, 9, 651317. [Google Scholar] [CrossRef]
  143. Chen, G.; Zhang, J.; Fu, Q.; Taly, V.; Tan, F. Integrative Analysis of Multi-Omics Data for Liquid Biopsy. Br. J. Cancer 2022, 128, 505–518. [Google Scholar] [CrossRef]
  144. Xu, H.; Lien, T.; Bergholtz, H.; Fleischer, T.; Djerroudi, L.; Vincent-Salomon, A.; Sørlie, T.; Aittokallio, T. Multi-Omics Marker Analysis Enables Early Prediction of Breast Tumor Progression. Front. Genet. 2021, 12, 670749. [Google Scholar] [CrossRef]
  145. Cowell, C.F.; Weigelt, B.; Sakr, R.A.; Ng, C.K.Y.; Hicks, J.; King, T.A.; Reis-Filho, J.S. Progression from Ductal Carcinoma in Situ to Invasive Breast Cancer: Revisited. Mol. Oncol. 2013, 7, 859–869. [Google Scholar] [CrossRef] [PubMed]
  146. Ren, H.; Liu, X.; Li, F.; He, X.; Zhao, N. Identification of a Six Gene Prognosis Signature for Papillary Thyroid Cancer Using Multi-Omics Methods and Bioinformatics Analysis. Front. Oncol. 2021, 11, 624421. [Google Scholar] [CrossRef]
  147. Piga, I.; L’Imperio, V.; Capitoli, G.; Denti, V.; Smith, A.; Magni, F.; Pagni, F. Paving the Path toward Multi-Omics Approaches in the Diagnostic Challenges Faced in Thyroid Pathology. Expert Rev. Proteom. 2023, 20, 419–437. [Google Scholar] [CrossRef] [PubMed]
  148. Ke, Y.; Ge, W. Identification of Prognostic Biomarkers in Neuroblastoma Using WGCNA and Multi-Omics Analysis. Discover Oncol. 2024, 15, 469. [Google Scholar] [CrossRef] [PubMed]
  149. Deng, C.; Xie, C.; Li, Z.; Mei, J.; Wang, K. Multi-Omics Analysis Identifies Diagnostic Circulating Biomarkers and Potential Therapeutic Targets, Revealing IQGAP1 as an Oncogene in Gastric Cancer. NPJ Precis. Oncol. 2025, 9, 1–18. [Google Scholar] [CrossRef]
  150. Kellogg, R.A.; Dunn, J.; Snyder, M.P. Personal Omics for Precision Health. Circ. Res. 2018, 122, 1169–1171. [Google Scholar] [CrossRef]
  151. Chen, R.; Mias, G.I.; Li-Pook-Than, J.; Jiang, L.; Lam, H.Y.K.; Chen, R.; Miriami, E.; Karczewski, K.J.; Hariharan, M.; Dewey, F.E.; et al. Personal Omics Profiling Reveals Dynamic Molecular and Medical Phenotypes. Cell 2012, 148, 1293–1307. [Google Scholar] [CrossRef]
  152. Metwally, A.A.; Zhang, T.; Wu, S.; Kellogg, R.; Zhou, W.; Contrepois, K.; Tang, H.; Snyder, M. Robust Identification of Temporal Biomarkers in Longitudinal Omics Studies. Bioinformatics 2022, 38, 3802–3811. [Google Scholar] [CrossRef]
  153. Luo, J.; Shen, L.; Zheng, D. Diagnostic Value of Circulating Free DNA for the Detection of EGFR Mutation Status in NSCLC: A Systematic Review and Meta-Analysis. Sci. Rep. 2014, 4, 1–7. [Google Scholar] [CrossRef]
  154. Palmieri, M.; Zulato, E.; Wahl, S.G.F.; Guibert, N.; Frullanti, E. Diagnostic Accuracy of Circulating Free DNA Testing for the Detection of KRAS Mutations in Non-Small Cell Lung Cancer: A Systematic Review and Meta-Analysis. Front. Genet. 2022, 13, 1015161. [Google Scholar] [CrossRef]
  155. Tepus, M.; Yau, T.O. Non-Invasive Colorectal Cancer Screening: An Overview. Gastrointest. Tumors 2020, 7, 62–73. [Google Scholar] [CrossRef]
  156. Bardol, T.; Dujon, A.M.; Taly, V.; Dunyach-Remy, C.; Lavigne, J.P.; Costa-Silva, B.; Kurma, K.; Eslami-S, Z.; Cayrefourcq, L.; Canivet, C.; et al. Early Detection of Pancreatic Cancer by Liquid Biopsy “PANLIPSY”: A French Nation-Wide Study Project. BMC Cancer 2024, 24, 1–10. [Google Scholar] [CrossRef] [PubMed]
  157. Cristiano, S.; Leal, A.; Phallen, J.; Fiksel, J.; Adleff, V.; Bruhm, D.C.; Jensen, S.Ø.; Medina, J.E.; Hruban, C.; White, J.R.; et al. Genome-Wide Cell-Free DNA Fragmentation in Patients with Cancer. Nature 2019, 570, 385–389. [Google Scholar] [CrossRef] [PubMed]
  158. Klein, E.A.; Richards, D.; Cohn, A.; Tummala, M.; Lapham, R.; Cosgrove, D.; Chung, G.; Clement, J.; Gao, J.; Hunkapiller, N.; et al. Clinical Validation of a Targeted Methylation-Based Multi-Cancer Early Detection Test Using an Independent Validation Set. Ann. Oncol. 2021, 32, 1167–1177. [Google Scholar] [CrossRef]
  159. Schrag, D.; McDonnell, C.H.; Nadauld, L.; Dilaveri, C.A.; Klein, E.A.; Reid, R.; Marinac, C.R.; Chung, K.C.; Lopatin, M.; Fung, E.T.; et al. A Prospective Study of a Multi-Cancer Early Detection Blood Test. Ann. Oncol. 2022, 33, S961. [Google Scholar] [CrossRef]
  160. Bratulic, S.; Limeta, A.; Dabestani, S.; Birgisson, H.; Enblad, G.; Stålberg, K.; Hesselager, G.; Häggman, M.; Höglund, M.; Simonson, O.E.; et al. Noninvasive Detection of Any-Stage Cancer Using Free Glycosaminoglycans. Proc. Natl. Acad. Sci. USA 2022, 119, e2115328119. [Google Scholar] [CrossRef]
  161. Cohen, J.D.; Li, L.; Wang, Y.; Thoburn, C.; Afsari, B.; Danilova, L.; Douville, C.; Javed, A.A.; Wong, F.; Mattox, A.; et al. Detection and Localization of Surgically Resectable Cancers with a Multi-Analyte Blood Test. Science 2018, 359, 926–930. [Google Scholar] [CrossRef]
  162. Rolfo, C.; Russo, A. Liquid Biopsy for Early Stage Lung Cancer Moves Ever Closer. Nat. Rev. Clin. Oncol. 2020, 17, 523–524. [Google Scholar] [CrossRef]
  163. Abbosh, C.; Birkbak, N.J.; Swanton, C. Early Stage NSCLC—Challenges to Implementing CtDNA-Based Screening and MRD Detection. Nat. Rev. Clin. Oncol. 2018, 15, 9. [Google Scholar] [CrossRef]
  164. Rolfo, C.; Mack, P.; Scagliotti, G.V.; Aggarwal, C.; Arcila, M.E.; Barlesi, F.; Bivona, T.; Diehn, M.; Dive, C.; Dziadziuszko, R.; et al. Liquid Biopsy for Advanced NSCLC: A Consensus Statement From the International Association for the Study of Lung Cancer. J. Thorac. Oncol. 2021, 16, 1647–1662. [Google Scholar] [CrossRef]
  165. Mathios, D.; Johansen, J.S.; Cristiano, S.; Medina, J.E.; Phallen, J.; Larsen, K.R.; Bruhm, D.C.; Niknafs, N.; Ferreira, L.; Adleff, V.; et al. Detection and Characterization of Lung Cancer Using Cell-Free DNA Fragmentomes. Nat. Commun. 2021, 12, 5060. [Google Scholar] [CrossRef] [PubMed]
  166. Singh, S.; Podder, P.S.; Russo, M.; Henry, C.; Cinti, S. Tailored Point-of-Care Biosensors for Liquid Biopsy in the Field of Oncology. Lab Chip 2022, 23, 44–61. [Google Scholar] [CrossRef] [PubMed]
  167. Fan, Z.; Zhou, Y.; Ressom, H.W. MOTA: Network-Based Multi-Omic Data Integration for Biomarker Discovery. Metabolites 2020, 10, 144. [Google Scholar] [CrossRef]
  168. Lan, W.; Liao, H.; Chen, Q.; Zhu, L.; Pan, Y.; Chen, Y.P.P. DeepKEGG: A Multi-Omics Data Integration Framework with Biological Insights for Cancer Recurrence Prediction and Biomarker Discovery. Brief. Bioinform. 2024, 25, bbae185. [Google Scholar] [CrossRef]
  169. Shi, K.; Lin, W.; Zhao, X.M. Identifying Molecular Biomarkers for Diseases with Machine Learning Based on Integrative Omics. Trans. Comput. Biol. Bioinform. 2021, 18, 2514–2525. [Google Scholar] [CrossRef]
  170. Ozaki, Y.; Broughton, P.; Abdollahi, H.; Valafar, H.; Blenda, A.V.; Lu, L.; Kumar, A.; Ozaki, Y.; Broughton, P.; Abdollahi, H.; et al. Integrating Omics Data and AI for Cancer Diagnosis and Prognosis. Cancers 2024, 16, 2448. [Google Scholar] [CrossRef] [PubMed]
  171. Guan, X.; Du, Y.; Ma, R.; Teng, N.; Ou, S.; Zhao, H.; Li, X. Construction of the XGBoost Model for Early Lung Cancer Prediction Based on Metabolic Indices. BMC Med. Inform. Decis. Mak. 2023, 23, 1–16. [Google Scholar] [CrossRef]
  172. Dessie, E.Y.; Tsai, J.J.P.; Chang, J.G.; Ng, K.L. A Novel MiRNA-Based Classification Model of Risks and Stages for Clear Cell Renal Cell Carcinoma Patients. BMC Bioinform. 2021, 22, 1–16. [Google Scholar] [CrossRef]
  173. Goswami, C.; Chawla, S.; Thakral, D.; Pant, H.; Verma, P.; Malik, P.S.; Jayadeva; Gupta, R.; Ahuja, G.; Sengupta, D. Molecular Signature Comprising 11 Platelet-Genes Enables Accurate Blood-Based Diagnosis of NSCLC. BMC Genom. 2020, 21, 1–12. [Google Scholar] [CrossRef]
  174. Gupta, R.; Kleinjans, J.; Caiment, F. Identifying Novel Transcript Biomarkers for Hepatocellular Carcinoma (HCC) Using RNA-Seq Datasets and Machine Learning. BMC Cancer 2021, 21, 1–15. [Google Scholar] [CrossRef]
  175. Tran, Q.T.; Alom, M.Z.; Orr, B.A. Comprehensive Study of Semi-Supervised Learning for DNA Methylation-Based Supervised Classification of Central Nervous System Tumors. BMC Bioinform. 2022, 23, 1–17. [Google Scholar] [CrossRef]
  176. Tao, K.; Bian, Z.; Zhang, Q.; Guo, X.; Yin, C.; Wang, Y.; Zhou, K.; Wan, S.; Shi, M.; Bao, D.; et al. Machine Learning-Based Genome-Wide Interrogation of Somatic Copy Number Aberrations in Circulating Tumor DNA for Early Detection of Hepatocellular Carcinoma. EBioMedicine 2020, 56, 102811. [Google Scholar] [CrossRef]
  177. Shaukat, A.; Burke, C.A.; Chan, A.T.; Grady, W.M.; Gupta, S.; Katona, B.W.; Ladabaum, U.; Liang, P.S.; Liu, J.J.; Putcha, G.; et al. Clinical Validation of a Circulating Tumor DNA–Based Blood Test to Screen for Colorectal Cancer. JAMA 2025, 334, 56–63. [Google Scholar] [CrossRef]
  178. Putcha, G.; Xu, C.; Shaukat, A.; Levin, T.R. Prevention of colorectal cancer through multiomics blood testing: The PREEMPT CRC study. J. Clin. Oncol. 2022, 2022, 40. [Google Scholar] [CrossRef]
  179. Lin, J.; Ariazi, E.; Dzamba, M.; Hsu, T.-K.; Kothen-Hill, S.; Li, K.; Liu, T.-Y.; Mahajan, S.; Palaniappan, K.K.; Pasupathy, A.; et al. Evaluation of a sensitive blood test for the detection of colorectal advanced adenomas in a prospective cohort using a multiomics approach. J. Clin. Oncol. 2021, 39, 43. [Google Scholar] [CrossRef]
  180. Nagarkar, R.; Gopichand, M.; Pal, S.K.; Gupta, A.; Saquib, N.M.; Ahmad, A.; Sagar, G.; Rao, K.V.S.; Siddiqui, Z.; Longkumer, I. Development of a Serum Metabolome-Based Test for Early-Stage Detection of Multiple Cancers. Cancer Rep. 2024, 7, e70042. [Google Scholar] [CrossRef]
  181. Papier, K.; Atkins, J.R.; Tong, T.Y.N.; Gaitskell, K.; Desai, T.; Ogamba, C.F.; Parsaeian, M.; Reeves, G.K.; Mills, I.G.; Key, T.J.; et al. Identifying Proteomic Risk Factors for Cancer Using Prospective and Exome Analyses of 1463 Circulating Proteins and Risk of 19 Cancers in the UK Biobank. Nat. Commun. 2024, 15, 4010. [Google Scholar] [CrossRef]
  182. Harlid, S.; Myte, R.; Van Guelpen, B. The Metabolic Syndrome, Inflammation, and Colorectal Cancer Risk: An Evaluation of Large Panels of Plasma Protein Markers Using Repeated, Prediagnostic Samples. Mediat. Inflamm. 2017, 2017, 4803156. [Google Scholar] [CrossRef]
  183. Sun, X.; Shu, X.O.; Lan, Q.; Laszkowska, M.; Cai, Q.; Rothman, N.; Wen, W.; Zheng, W.; Shu, X. Prospective Proteomic Study Identifies Potential Circulating Protein Biomarkers for Colorectal Cancer Risk. Cancers 2022, 14, 3261. [Google Scholar] [CrossRef]
  184. Cominetti, O.; Dayon, L. Unravelling Disease Complexity: Integrative Analysis of Multi-Omic Data in Clinical Research. Expert Rev. Proteom. 2025, 22, 149–162. [Google Scholar] [CrossRef]
  185. Bhatt, A.A.; Niell, B. Tumor Doubling Time and Screening Interval. Radiol. Clin. N. Am. 2024, 62, 571–580. [Google Scholar] [CrossRef]
  186. Kim, J.; Campbell, A.S.; de Ávila, B.E.F.; Wang, J. Wearable Biosensors for Healthcare Monitoring. Nat. Biotechnol. 2019, 37, 389–406. [Google Scholar] [CrossRef]
  187. Heikenfeld, J.; Jajack, A.; Rogers, J.; Gutruf, P.; Tian, L.; Pan, T.; Li, R.; Khine, M.; Kim, J.; Wang, J.; et al. Wearable Sensors: Modalities, Challenges, and Prospects. Lab Chip 2018, 18, 217–248. [Google Scholar] [CrossRef]
  188. Mahato, K.; Saha, T.; Ding, S.; Sandhu, S.S.; Chang, A.Y.; Wang, J. Hybrid Multimodal Wearable Sensors for Comprehensive Health Monitoring. Nat. Electron. 2024, 7, 735–750. [Google Scholar] [CrossRef]
  189. Coskun, A.; Savas, I.N.; Can, O.; Lippi, G. From Population-Based to Personalized Laboratory Medicine: Continuous Monitoring of Individual Laboratory Data with Wearable Biosensors. Crit. Rev. Clin. Lab. Sci. 2025, 62, 198–227. [Google Scholar] [CrossRef]
  190. Rum, L.; Sten, O.; Vendrame, E.; Belluscio, V.; Camomilla, V.; Vannozzi, G.; Truppa, L.; Notarantonio, M.; Sciarra, T.; Lazich, A.; et al. Wearable Sensors in Sports for Persons with Disability: A Systematic Review. Sensors 2021, 21, 1858–2021. [Google Scholar] [CrossRef]
  191. Olmedo-Aguirre, J.O.; Reyes-Campos, J.; Alor-Hernández, G.; Machorro-Cano, I.; Rodríguez-Mazahua, L.; Sánchez-Cervantes, J.L. Remote Healthcare for Elderly People Using Wearables: A Review. Biosensors 2022, 12, 73. [Google Scholar] [CrossRef]
  192. Sohrabi, H.; Bolandi, N.; Hemmati, A.; Eyvazi, S.; Ghasemzadeh, S.; Baradaran, B.; Oroojalian, F.; Reza Majidi, M.; de la Guardia, M.; Mokhtarzadeh, A. State-of-the-Art Cancer Biomarker Detection by Portable (Bio) Sensing Technology: A Critical Review. Microchem. J. 2022, 177, 107248. [Google Scholar] [CrossRef]
  193. Qiu, Z.; Shu, J.; Tang, D. Bioresponsive Release System for Visual Fluorescence Detection of Carcinoembryonic Antigen from Mesoporous Silica Nanocontainers Mediated Optical Color on Quantum Dot-Enzyme-Impregnated Paper. Anal. Chem. 2017, 89, 5152–5160. [Google Scholar] [CrossRef]
  194. Chi, L.; Wang, X.; Chen, H.; Tang, D.; Xue, F. Paper-Based Photoelectrochemical Immunoassay for Ultrasensitive Screening of Carcinoembryonic Antigen on Hollow CdS/CdMoO4-Functionalized Photoanode. Talanta 2023, 254, 124176. [Google Scholar] [CrossRef]
  195. Tsogka, I.; Mermiga, E.; Pagkali, V.; Kokkinos, C.; Economou, A. A Simplified Lateral Flow Immunosensor for the Assay of Carcinoembryonic Antigen in Low-Resource Settings. Anal. Methods 2024, 16, 2921–2929. [Google Scholar] [CrossRef]
  196. Gu, Y.; Li, Z.; Ge, S.; Mao, Y.; Gu, Y.; Cao, X.; Lu, D. A Microfluidic Chip Using Au@SiO2 Array–Based Highly SERS-Active Substrates for Ultrasensitive Detection of Dual Cervical Cancer–Related Biomarkers. Anal. Bioanal. Chem. 2022, 414, 7659–7673. [Google Scholar] [CrossRef]
  197. Ibáñez-Redín, G.; Materon, E.M.; Furuta, R.H.M.; Wilson, D.; do Nascimento, G.F.; Melendez, M.E.; Carvalho, A.L.; Reis, R.M.; Oliveira, O.N.; Gonçalves, D. Screen-Printed Electrodes Modified with Carbon Black and Polyelectrolyte Films for Determination of Cancer Marker Carbohydrate Antigen 19-9. Microchim. Acta 2020, 187, 1–11. [Google Scholar] [CrossRef]
  198. Huang, Y.; Wen, Y.; Baryeh, K.; Takalkar, S.; Lund, M.; Zhang, X.; Liu, G. Lateral Flow Assay for Carbohydrate Antigen 19–9 in Whole Blood by Using Magnetized Carbon Nanotubes. Microchim. Acta 2017, 184, 4287–4294. [Google Scholar] [CrossRef]
  199. Tripathi, P.; Kumar, A.; Sachan, M.; Gupta, S.; Nara, S. Aptamer-Gold Nanozyme Based Competitive Lateral Flow Assay for Rapid Detection of CA125 in Human Serum. Biosens. Bioelectron. 2020, 165, 112368. [Google Scholar] [CrossRef]
  200. Fan, Y.; Shi, S.; Ma, J.; Guo, Y. Smartphone-Based Electrochemical System with Multi-Walled Carbon Nanotubes/Thionine/Gold Nanoparticles Modified Screen-Printed Immunosensor for Cancer Antigen 125 Detection. Microchem. J. 2022, 174, 107044. [Google Scholar] [CrossRef]
  201. Rebelo, T.S.C.R.; Ribeiro, J.A.; Sales, M.G.F.; Pereira, C.M. Electrochemical Immunosensor for Detection of CA 15-3 Biomarker in Point-of-Care. Sens. Biosensing Res. 2021, 33, 100445. [Google Scholar] [CrossRef]
  202. Cui, C.; Xu, M.; Guan, Q.; Wang, X.; Yang, G.; Zhang, Z.; Du, J. Electrochemical Detection of Alpha-Fetoprotein Using Graphene-Assisted Sensors: A Novel Approach for Liver Cancer Screening. Alex. Eng. J. 2025, 123, 511–518. [Google Scholar] [CrossRef]
  203. Yu, Z.; Huang, L.; Chen, J.; Li, M.; Tang, D. Graded Oxygen-Doped CdS Electrode for Portable Photoelectrochemical Immunoassay of Alpha-Fetoprotein Coupling with a Digital Multimeter Readout. Sens. Actuators B Chem. 2021, 343, 130136. [Google Scholar] [CrossRef]
  204. Jia, Y.; Zhao, H.; Huang, S.; Yin, F.; Wang, W.; Hu, Q.; Wang, Y.; Feng, B. Rapid and Portable Detection of Hepatocellular Carcinoma Marker Alpha-Fetoprotein Using a Droplet Evaporation-Based Biosensor. Talanta 2025, 294, 128189. [Google Scholar] [CrossRef]
  205. He, W.; Liu, L.; Cao, Z.; Lin, Y.; Tian, Y.; Zhang, Q.; Zhou, C.; Ye, X.; Cui, T. Shrink Polymer Based Electrochemical Sensor for Point-of-Care Detection of Prostate-Specific Antigen. Biosens. Bioelectron. 2023, 228, 115193. [Google Scholar] [CrossRef]
  206. Sena-Torralba, A.; Banguera-Ordoñez, Y.D.; Carrascosa, J.; Maquieira, Á.; Morais, S. Portable Electrophoretic Lateral Flow Biosensing for Ultra-Sensitive Human Lactate Dehydrogenase Detection in Serum Samples. Biosens. Bioelectron. 2025, 282, 117504. [Google Scholar] [CrossRef]
  207. Banguera-Ordoñez, Y.D.; Sena-Torralba, A.; Quintero-Campos, P.; Maquieira, Á.; Morais, S. Smartphone-Based Lateral Flow Immunoassay for Sensitive Determination of Lactate Dehydrogenase at the Point of Care. Talanta 2025, 281, 126803. [Google Scholar] [CrossRef]
  208. Kim, H.M.; Jeong, D.H.; Lee, H.Y.; Park, J.H.; Lee, S.K. Design and Validation of Fiber Optic Localized Surface Plasmon Resonance Sensor for Thyroglobulin Immunoassay with High Sensitivity and Rapid Detection. Sci. Rep. 2021, 11, 1–9. [Google Scholar] [CrossRef]
  209. Xu, J.; Chen, H.; Luo, X.; Guo, B.; Jia, L.; Wang, F. Internal Three-Dimensional Graphdiyne-Based Self-Powered Biosensor Integrated with External Physical Power for Portable Detection of Tumor Markers. Sens. Actuators B Chem. 2025, 426, 137114. [Google Scholar] [CrossRef]
  210. Srilikhit, A.; Kongkeaw, S.; Cotchim, S.; Janduang, S.; Wannapob, R.; Kanatharana, P.; Thavarungkul, P.; Limbut, W. Carcinoembryonic Antigen and Cancer Antigen 125 Simultaneously Determined Using a Fluidics-Integrated Dual Carbon Electrode. Microchem. J. 2024, 204, 110911. [Google Scholar] [CrossRef]
  211. Chu, T.; Wang, H.; Qiu, Y.; Luo, H.; He, B.; Wu, B.; Gao, B. 3D Printed Smart Silk Wearable Sensors. Analyst 2021, 146, 1552–1558. [Google Scholar] [CrossRef]
  212. Barbosa, A.I.; Gehlot, P.; Sidapra, K.; Edwards, A.D.; Reis, N.M. Portable Smartphone Quantitation of Prostate Specific Antigen (PSA) in a Fluoropolymer Microfluidic Device. Biosens. Bioelectron. 2015, 70, 5–14. [Google Scholar] [CrossRef]
  213. Chen, C.H.; Wang, E.; Lee, T.-H.; Huang, C.-C.; Tai, C.-S.; Lin, Y.-R.; Chen, W.-L. Point-of-Care NSE Biosensor for Objective Assessment of Stroke Risk. Biosensors 2025, 15, 264. [Google Scholar] [CrossRef]
  214. Zhang, S.; Luo, Y.; Zhuang, W.; Zhong, G.; Su, L.; Xu, T.; Zhang, X. Fully Integrated Ratiometric Fluorescence Enrichment Platform for High-Sensitivity POC Testing of Salivary Cancer Biomarkers. Anal. Chem. 2023, 95, 18739–18747. [Google Scholar] [CrossRef]
  215. Zhou, S.; Tu, D.; Liu, Y.; You, W.; Zhang, Y.; Zheng, W.; Chen, X. Ultrasensitive Point-of-Care Test for Tumor Marker in Human Saliva Based on Luminescence-Amplification Strategy of Lanthanide Nanoprobes. Adv. Sci. 2021, 8, 2002657. [Google Scholar] [CrossRef]
  216. Tofighi, F.B.; Saadati, A.; Kholafazad-kordasht, H.; Farshchi, F.; Hasanzadeh, M.; Samiei, M. Electrochemical Immunoplatform to Assist in the Diagnosis of Oral Cancer through the Determination of CYFRA 21.1 Biomarker in Human Saliva Samples: Preparation of a Novel Portable Biosensor toward Non-Invasive Diagnosis of Oral Cancer. J. Mol. Recognit. 2021, 34, e2932. [Google Scholar] [CrossRef]
  217. Joshi, S.; Kallappa, S.; Kumar, P.; Shukla, S.; Ghosh, R. Simple Diagnosis of Cancer by Detecting CEA and CYFRA 21-1 in Saliva Using Electronic Sensors. Sci. Rep. 2022, 12, 1–13. [Google Scholar] [CrossRef]
  218. Oliveira, A.E.F.; Pereira, A.C.; Resende, M.A.C.; Ferreira, L.F. Disposable Voltammetric Immunosensor for Determination and Quantification of Biomarker CA 15-3 in Biological Specimens. Analytica 2024, 5, 74–89. [Google Scholar] [CrossRef]
  219. Yang, Y.; Wang, J.; Huang, W.; Wan, G.; Xia, M.; Chen, D.; Zhang, Y.; Wang, Y.; Guo, F.; Tan, J.; et al. Integrated Urinalysis Devices Based on Interface-Engineered Field-Effect Transistor Biosensors Incorporated With Electronic Circuits. Adv. Mater. 2022, 34, 2203224. [Google Scholar] [CrossRef]
  220. Kim, S.; Kim, T.G.; Lee, S.H.; Kim, W.; Bang, A.; Moon, S.W.; Song, J.; Shin, J.H.; Yu, J.S.; Choi, S. Label-Free Surface-Enhanced Raman Spectroscopy Biosensor for On-Site Breast Cancer Detection Using Human Tears. ACS Appl. Mater. Interfaces 2020, 12, 7897–7904. [Google Scholar] [CrossRef]
  221. Nhu, C.T.; Do Quang, L.; Jen, C.P.; Duc, T.C.; Bui, T.T.; Vu Ngoc, T. NSE Protein Detection in a Microfluidic Channel Integrated an Electrochemical Biosensor. Biomed. Phys. Eng. Express 2024, 11, 015047. [Google Scholar] [CrossRef]
  222. Chen, H.; Chen, C.; Bai, S.; Gao, Y.; Metcalfe, G.; Cheng, W.; Zhu, Y. Multiplexed Detection of Cancer Biomarkers Using a Microfluidic Platform Integrating Single Bead Trapping and Acoustic Mixing Techniques. Nanoscale 2018, 10, 20196–20206. [Google Scholar] [CrossRef]
  223. Oliveira, D.; Romaguera Barcelay, Y.; Moreira, F.T.C. An Electrochemically Synthesized Molecularly Imprinted Polymer for Highly Selective Detection of Breast Cancer Biomarker CA 15-3: A Promising Point-of-Care Biosensor. RSC Adv. 2024, 14, 15347–15357. [Google Scholar] [CrossRef]
  224. Heikenfeld, J.; Jajack, A.; Feldman, B.; Granger, S.W.; Gaitonde, S.; Begtrup, G.; Katchman, B.A. Accessing Analytes in Biofluids for Peripheral Biochemical Monitoring. Nat. Biotechnol. 2019, 37, 407–419. [Google Scholar] [CrossRef]
  225. Zhang, Z.; Li, Z.; Wei, K.; Cao, Z.; Zhu, Z.; Chen, R. Sweat as a Source of Non-Invasive Biomarkers for Clinical Diagnosis: An Overview. Talanta 2024, 273, 125865. [CrossRef]
  226. Mohankumar, P.; Ajayan, J.; Mohanraj, T.; Yasodharan, R. Recent Developments in Biosensors for Healthcare and Biomedical Applications: A Review. Measurement 2021, 167, 108293. [Google Scholar] [CrossRef]
  227. Jayanthi, V.S.P.K.S.A.; Das, A.B.; Saxena, U. Recent Advances in Biosensor Development for the Detection of Cancer Biomarkers. Biosens. Bioelectron. 2017, 91, 15–23. [Google Scholar] [CrossRef]
  228. Hroncekova, S.; Bertok, T.; Hires, M.; Jane, E.; Lorencova, L.; Vikartovska, A.; Tanvir, A.; Kasak, P.; Tkac, J. Ultrasensitive Ti3C2TX MXene/Chitosan Nanocomposite-Based Amperometric Biosensor for Detection of Potential Prostate Cancer Marker in Urine Samples. Processes 2020, 8, 580. [Google Scholar] [CrossRef]
  229. Zhang, S.; Rong, F.; Guo, C.; Duan, F.; He, L.; Wang, M.; Zhang, Z.; Kang, M.; Du, M. Metal–Organic Frameworks (MOFs) Based Electrochemical Biosensors for Early Cancer Diagnosis in Vitro. Coord. Chem. Rev. 2021, 439, 213948. [Google Scholar] [CrossRef]
  230. Kappen, J.; Skorupa, M.; Krukiewicz, K. Conducting Polymers as Versatile Tools for the Electrochemical Detection of Cancer Biomarkers. Biosensors 2023, 13, 31. [Google Scholar] [CrossRef]
  231. Afshari Babazad, M.; Foroozandeh, A.; Abdouss, M.; SalarAmoli, H.; Babazad, R.A.; Hasanzadeh, M. Recent Progress and Challenges in Biosensing of Carcinoembryonic Antigen. TrAC 2024, 180, 117964. [Google Scholar] [CrossRef]
  232. Khan, H.; Shah, M.R.; Barek, J.; Malik, M.I. Cancer Biomarkers and Their Biosensors: A Comprehensive Review. TrAC 2023, 158, 116813. [Google Scholar] [CrossRef]
  233. Sun, S.; Chen, J. Recent Advances in Hydrogel-Based Biosensors for Cancer Detection. ACS Appl. Mater. Interfaces 2024, 16, 46988–47002. [Google Scholar] [CrossRef]
  234. Gu, X.; She, Z.; Ma, T.; Tian, S.; Kraatz, H.B. Electrochemical Detection of Carcinoembryonic Antigen. Biosens. Bioelectron. 2018, 102, 610–616. [Google Scholar] [CrossRef]
  235. Mavrikou, S.; Moschopoulou, G.; Zafeirakis, A.; Kalogeropoulou, K.; Giannakos, G.; Skevis, A.; Kintzios, S. An Ultra-Rapid Biosensory Point-of-Care (POC) Assay for Prostate-Specific Antigen (PSA) Detection in Human Serum. Sensors 2018, 18, 3834. [Google Scholar] [CrossRef]
  236. Liu, Z.; Zhou, Y.; Lu, J.; Gong, T.; Ibáñez, E.; Cifuentes, A.; Lu, W. Microfluidic Biosensors for Biomarker Detection in Body Fluids: A Key Approach for Early Cancer Diagnosis. Biomark. Res. 2024, 12, 1–29. [Google Scholar] [CrossRef]
  237. Le, L.T.P.; Nguyen, A.H.Q.; Phan, L.M.T.; Ngo, H.T.T.; Wang, X.; Cunningham, B.; Valera, E.; Bashir, R.; Taylor-Robinson, A.W.; Do, C.D. Current Smartphone-Assisted Point-of-Care Cancer Detection: Towards Supporting Personalized Cancer Monitoring. TrAC 2024, 174, 117681. [Google Scholar] [CrossRef]
  238. Chanarsa, S.; Jakmunee, J.; Ounnunkad, K. A Sandwich-like Configuration with a Signal Amplification Strategy Using a Methylene Blue/Aptamer Complex on a Heterojunction 2D MoSe2/2D WSe2 Electrode: Toward a Portable and Sensitive Electrochemical Alpha-Fetoprotein Immunoassay. Front. Cell. Infect. Microbiol. 2022, 12, 916357. [Google Scholar] [CrossRef]
  239. Foroozandeh, A.; Babazad, M.A.; Jouybar, S.; Abdouss, M.; Salar Amoli, H.; Dashtian, K.; Hasanzadeh, M. Recent Advancements in Biosensors for Diagnosis of Ovarian Cancer: Analytical Approaches. TrAC 2025, 183, 118119. [Google Scholar] [CrossRef]
  240. Wang, L.; Balasubramanian, P.; Chen, A.P.; Kummar, S.; Evrard, Y.A.; Kinders, R.J. Promise and Limits of the CellSearch Platform for Evaluating Pharmacodynamics in Circulating Tumor Cells. Semin. Oncol. 2016, 43, 464–475. [Google Scholar] [CrossRef]
  241. Dizdar, L.; Fluegen, G.; van Dalum, G.; Honisch, E.; Neves, R.P.; Niederacher, D.; Neubauer, H.; Fehm, T.; Rehders, A.; Krieg, A.; et al. Detection of Circulating Tumor Cells in Colorectal Cancer Patients Using the GILUPI CellCollector: Results from a Prospective, Single-Center Study. Mol. Oncol. 2019, 13, 1548–1558. [Google Scholar] [CrossRef]
  242. Dong, Y.; Mao, S.; Chen, S.; Ma, J.; Jaffrezic-Renault, N.; Guo, Z. Opportunities and challenges of microneedle electrochemical sensors for interstitial fluid detection. TrAC 2024, 180, 117891. [Google Scholar] [CrossRef]
  243. Miller, P.R.; Taylor, R.M.; Tran, B.Q.; Boyd, G.; Glaros, T.; Chavez, V.H.; Krishnakumar, R.; Sinha, A.; Poorey, K.; Williams, K.P.; et al. Extraction and Biomolecular Analysis of Dermal Interstitial Fluid Collected with Hollow Microneedles. Commun. Biol. 2018, 1, 173. [Google Scholar] [CrossRef]
  244. Merzougui, C.; Yang, X.; Meng, D.; Huang, Y.; Zhao, X. Microneedle Array-Based Dermal Interstitial Fluid Biopsy for Cancer Diagnosis: Advances and Challenges. Adv. Healthc. Mater. 2025, 14, 2404420. [Google Scholar] [CrossRef]
  245. Huang, H.; Qu, M.; Zhou, Y.; Cao, W.; Huang, X.; Sun, J.; Sun, W.; Zhou, X.; Xu, M.; Jiang, X. A Microneedle Patch for Breast Cancer Screening via Minimally Invasive Interstitial Fluid Sampling. Chem. Eng. J. 2023, 472, 145036. [Google Scholar] [CrossRef]
  246. Maqsood, Q.; Sumrin, A.; Saleem, Y.; Wajid, A.; Mahnoor, M. Exosomes in Cancer: Diagnostic and Therapeutic Applications. Clin. Med. Insights Oncol. 2024, 18, 11795549231215966. [Google Scholar] [CrossRef]
  247. Park, W.; Maeng, S.W.; Mok, J.W.; Choi, M.; Cha, H.J.; Joo, C.K.; Hahn, S.K. Hydrogel Microneedles Extracting Exosomes for Early Detection of Colorectal Cancer. Biomacromolecules 2023, 24, 1445–1452. [Google Scholar] [CrossRef]
  248. Bratei, A.A.; Stefan-van Staden, R.I.; Ilie-Mihai, R.M.; Gheorghe, D.C. Simultaneous Assay of CA 72-4, CA 19-9, CEA and CA 125 in Biological Samples Using Needle Three-Dimensional Stochastic Microsensors. Sensors 2023, 23, 8046. [Google Scholar] [CrossRef]
  249. Liu, J.; Tang, Y.; Cheng, Y.; Huang, W.; Xiang, L. Electrochemical Biosensors Based on Saliva Electrolytes for Rapid Detection and Diagnosis. J. Mater. Chem. B 2022, 11, 33–54. [Google Scholar] [CrossRef]
  250. AlAli, A.M.; Walsh, T.; Maranzano, M. CYFRA 21-1 and MMP-9 as Salivary Biomarkers for the Detection of Oral Squamous Cell Carcinoma: A Systematic Review of Diagnostic Test Accuracy. Int. J. Oral Maxillofac. Surg. 2020, 49, 973–983. [Google Scholar] [CrossRef]
  251. Chen, M.; Zhao, H. Next-Generation Sequencing in Liquid Biopsy: Cancer Screening and Early Detection. Hum. Genom. 2019, 13, 34. [Google Scholar] [CrossRef]
  252. Jafari, M.; Hasanzadeh, M. Non-Invasive Bioassay of Cytokeratin Fragment 21.1 (Cyfra 21.1) Protein in Human Saliva Samples Using Immunoreaction Method: An Efficient Platform for Early-Stage Diagnosis of Oral Cancer Based on Biomedicine. Biomed. Pharmacother. 2020, 131, 110671. [Google Scholar] [CrossRef]
  253. Kumar, S.; Kumar, S.; Tiwari, S.; Augustine, S.; Srivastava, S.; Yadav, B.K.; Malhotra, B.D. Highly Sensitive Protein Functionalized Nanostructured Hafnium Oxide Based Biosensing Platform for Non-Invasive Oral Cancer Detection. Sens. Actuators B Chem. 2016, 235, 1–10. [Google Scholar] [CrossRef]
  254. Chan, K.M.; Gleadle, J.M.; O’Callaghan, M.; Vasilev, K.; MacGregor, M. Prostate Cancer Detection: A Systematic Review of Urinary Biosensors. Prostate Cancer Prostatic Dis. 2022, 25, 39–46. [Google Scholar] [CrossRef]
  255. Cao, X.; Gao, Y.; Ding, W.; Li, C. A Wearable Electrochemical Sensor for Testing Carcinoembryonic Antigen in Sweat. In Proceedings of the 2023 4th International Symposium on Artificial Intelligence for Medicine Science, Chengdu, China, 20–22 October 2023; pp. 722–727. [Google Scholar] [CrossRef]
  256. 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]
  257. Evans, V.; Vockler, C.; Friedlander, M.; Walsh, B.; Willcox, M.D.P. Lacryglobin in Human Tears, a Potential Marker for Cancer. Clin. Exp. Ophthalmol. 2001, 29, 161–163. [Google Scholar] [CrossRef]
  258. Daily, A.; Ravishankar, P.; Harms, S.; Klimberg, V.S. Using Tears as a Non-Invasive Source for Early Detection of Breast Cancer. PLoS ONE 2022, 17, e0267676. [Google Scholar] [CrossRef] [PubMed]
  259. Dikovskaya, M.A.; Trunov, A.N.; Chernykh, V.V.; Korolenko, T.A. Cystatin C and lactoferrin concentrations in biological fluids as possible prognostic factors in eye tumor development. Int. J. Circumpolar Health 2013, 72, 21087. [Google Scholar] [CrossRef] [PubMed]
  260. Ates, H.C.; Dincer, C. Wearable Breath Analysis. Nat. Rev. Bioeng. 2023, 1, 80–82. [Google Scholar] [CrossRef]
  261. Gashimova, E.; Temerdashev, A.; Perunov, D.; Porkhanov, V.; Polyakov, I. Diagnosis of Lung Cancer Through Exhaled Breath: A Comprehensive Study. Mol. Diagn. Ther. 2024, 28, 847–860. [Google Scholar] [CrossRef]
  262. VA, B.; Mathew, P.; Thomas, S.; Mathew, L. Detection of Lung Cancer and Stages via Breath Analysis Using a Self-Made Electronic Nose Device. Expert. Rev. Mol. Diagn. 2024, 24, 341–353. [Google Scholar] [CrossRef]
  263. Yu, Q.; Chen, J.; Fu, W.; Muhammad, K.G.; Li, Y.; Liu, W.; Xu, L.; Dong, H.; Wang, D.; Liu, J.; et al. Smartphone-Based Platforms for Clinical Detections in Lung-Cancer-Related Exhaled Breath Biomarkers: A Review. Biosensors 2022, 12, 223. [Google Scholar] [CrossRef]
  264. Bajtarevic, A.; Ager, C.; Pienz, M.; Klieber, M.; Schwarz, K.; Ligor, M.; Ligor, T.; Filipiak, W.; Denz, H.; Fiegl, M.; et al. Noninvasive detection of lung cancer by analysis of exhaled breath. BMC Cancer 2009, 9, 348. [Google Scholar] [CrossRef]
  265. Zompanti, A.; Finamore, P.; Longo, F.; Grasso, S.; Frasca, L.; Celoro, F.; Santonico, M.; Cenerini, C.; La Monica, L.; Sabatini, A.; et al. Sensor Technology Advancement Enhancing Exhaled Breath Portability: Device Set up and Pilot Test in the Longitudinal Study of Lung Cancer. Sens. Actuators B Chem. 2025, 423, 136735. [Google Scholar] [CrossRef]
  266. Picciariello, A.; Dezi, A.; Vincenti, L.; Spampinato, M.G.; Zang, W.; Riahi, P.; Scott, J.; Sharma, R.; Fan, X.; Altomare, D.F. Colorectal Cancer Diagnosis through Breath Test Using a Portable Breath Analyzer—Preliminary Data. Sensors 2024, 24, 2343. [Google Scholar] [CrossRef]
  267. Acevedo, D.; Carrillo Gómez, C.M.; Cuastumal Vasquez, J.K.; Ramos, C.A.; Prostate, J.; Manuel, C.; Carrillo Gómez, J.K.; Cuastumal Vasquez, C.A.; Ramos, J. Prostate Cancer Detection in Colombian Patients through E-Senses Devices in Exhaled Breath and Urine Samples. Chemosensors 2024, 12, 11. [Google Scholar] [CrossRef]
  268. Cao, Y.; Xia, J.; Li, L.; Zeng, Y.; Zhao, J.; Li, G. Electrochemical Biosensors for Cancer Diagnosis: Multitarget Analysis to Present Molecular Characteristics of Tumor Heterogeneity. JACS Au 2024, 4, 4655–4672. [Google Scholar] [CrossRef]
  269. Tang, L.; Chang, S.J.; Chen, C.J.; Liu, J.T. Non-Invasive Blood Glucose Monitoring Technology: A Review. Sensors 2020, 20, 6925. [Google Scholar] [CrossRef] [PubMed]
  270. Ye, C.; Wang, M.; Min, J.; Tay, R.Y.; Lukas, H.; Sempionatto, J.R.; Li, J.; Xu, C.; Gao, W. A Wearable Aptamer Nanobiosensor for Non-Invasive Female Hormone Monitoring. Nat. Nanotechnol. 2023, 19, 330–337. [Google Scholar] [CrossRef]
  271. Fang, L.; Ren, H.; Mao, X.; Zhang, S.; Cai, Y.; Xu, S.; Zhang, Y.; Li, L.; Ye, X.; Liang, B. Differential Amperometric Microneedle Biosensor for Wearable Levodopa Monitoring of Parkinson’s Disease. Biosensors 2022, 12, 102. [Google Scholar] [CrossRef] [PubMed]
  272. Goud, K.Y.; Moonla, C.; Mishra, R.K.; Yu, C.; Narayan, R.; Litvan, I.; Wang, J. Wearable Electrochemical Microneedle Sensor for Continuous Monitoring of Levodopa: Toward Parkinson Management. ACS Sens. 2019, 4, 2196–2204. [Google Scholar] [CrossRef]
  273. Tai, L.C.; Liaw, T.S.; Lin, Y.; Nyein, H.Y.Y.; Bariya, M.; Ji, W.; Hettick, M.; Zhao, C.; Zhao, J.; Hou, L.; et al. Wearable Sweat Band for Noninvasive Levodopa Monitoring. Nano Lett. 2019, 19, 6346–6351. [Google Scholar] [CrossRef] [PubMed]
  274. Wang, M.; Yang, Y.; Min, J.; Song, Y.; Tu, J.; Mukasa, D.; Ye, C.; Xu, C.; Heflin, N.; McCune, J.S.; et al. A Wearable Electrochemical Biosensor for the Monitoring of Metabolites and Nutrients. Nat. Biomed. Eng. 2022, 6, 1225–1235. [Google Scholar] [CrossRef]
  275. Du, W.; Zhang, L.; Suh, E.; Lin, D.; Marcus, C.; Ozkan, L.; Ahuja, A.; Fernandez, S.; Shuvo, I.I.; Sadat, D.; et al. Conformable Ultrasound Breast Patch for Deep Tissue Scanning and Imaging. Sci. Adv. 2023, 9, eadh5325. [Google Scholar] [CrossRef]
  276. Bhatia, D.; Paul, S.; Acharjee, T.; Ramachairy, S.S. Biosensors and Their Widespread Impact on Human Health. Sens. Int. 2024, 5, 100257. [Google Scholar] [CrossRef]
  277. Sheydaei, O.; Khajehsharifi, H.; Rajabi, H.R. Rapid and Selective Diagnose of Sarcosine in Urine Samples as Prostate Cancer Biomarker by Mesoporous Imprinted Polymeric Nanobeads Modified Electrode. Sens. Actuators B Chem. 2020, 309, 127559. [Google Scholar] [CrossRef]
  278. Anıl, İ.U.; Sezgintürk, M.K. MIP-Based Sensing Strategies for the Diagnosis of Prostate and Lung Cancers. Talanta Open 2025, 11, 100432. [Google Scholar] [CrossRef]
  279. Coskun, A.; Lippi, G. The Impact of Physiological Variations on Personalized Reference Intervals and Decision Limits: An in-Depth Analysis. Clin. Chem. Lab. Med. 2024, 62, 2140–2147. [Google Scholar] [CrossRef]
  280. Coskun, A.; Lippi, G. Personalized Laboratory Medicine in the Digital Health Era: Recent Developments and Future Challenges. Clin. Chem. Lab. Med. 2024, 62, 402–409. [Google Scholar] [CrossRef]
  281. Fraser, C.G. Biological Variation: From Principles to Practice; AACC Press: Washington, DC, USA, 2001. [Google Scholar]
  282. Sandberg, S.; Carobene, A.; Bartlett, B.; Coskun, A.; Fernandez-Calle, P.; Jonker, N.; Díaz-Garzón, J.; Aarsand, A.K. Biological Variation: Recent Development and Future Challenges. Clin. Chem. Lab. Med. 2023, 61, 741–750. [Google Scholar] [CrossRef]
  283. Petersen, P.H.; Fraser, C.G.; Sandberg, S.; Goldschmidt, H. The Index of Individuality Is Often a Misinterpreted Quantity Characteristic. Clin. Chem. Lab. Med. 1999, 37, 655–661. [Google Scholar] [CrossRef]
  284. Coşkun, A.; Sandberg, S.; Unsal, I.; Cavusoglu, C.; Serteser, M.; Kilercik, M.; Aarsand, A.K. Personalized and Population-Based Reference Intervals for 48 Common Clinical Chemistry and Hematology Measurands: A Comparative Study. Clin. Chem. 2023, 69, 1009–1030. [Google Scholar] [CrossRef] [PubMed]
  285. Coşkun, A.; Sandberg, S.; Unsal, I.; Cavusoglu, C.; Serteser, M.; Kilercik, M.; Aarsand, A.K. Personalized Reference Intervals in Laboratory Medicine: A New Model Based on Within-Subject Biological Variation. Clin. Chem. 2021, 67, 374–384. [Google Scholar] [CrossRef]
  286. Coskun, A.; Sandberg, S.; Unsal, I.; Serteser, M.; Aarsand, A.K. Personalized Reference Intervals: From Theory to Practice. Crit. Rev. Clin. Lab. Sci. 2022, 59, 501–516. [Google Scholar] [CrossRef]
  287. Coskun, A.; Sandberg, S.; Unsal, I.; Yavuz, F.G.; Cavusoglu, C.; Serteser, M.; Kilercik, M.; Aarsand, A.K. Personalized Reference Intervals—Statistical Approaches and Considerations. Clin. Chem. Lab. Med. 2022, 60, 629–635. [Google Scholar] [CrossRef] [PubMed]
  288. Coskun, A. Prediction Interval: A Powerful Statistical Tool for Monitoring Patients and Analytical Systems. Biochem. Med. 2024, 34, 020101. [Google Scholar] [CrossRef] [PubMed]
  289. Meeker, W.Q.; Hahn, G.J.; Escobar, L.A. Statistical Intervals: A Guide for Practitioners and Researchers; John Wiley & Sons: Hoboken, NJ, USA, 2017; ISBN 9780471687177. [Google Scholar]
  290. Geisser, S. Predictive Inference: An Introduction; Springer: Boston, MA, USA, 1993; ISBN 978-0-412-03471-8. [Google Scholar]
  291. Aarsand, A.K.; Fernandez-Calle, P.; Webster, C.; Coskun, A.; Gonzales-Lao, E.; Diaz-Garzon, J.; Jonker, N.; Simon, M.; Braga, F.; Perich, C.; et al. EFLM Biological Variation Database. Available online: https://biologicalvariation.eu (accessed on 1 May 2025).
  292. Roberts, W.L.; McMillin, G.A.; Burtis, C.A.; Bruns, D.E. Reference information for the clinical laboratory. In Tietz Textbook of Clinical Chemistry and Molecular Diagnostics, 5th ed.; Burtis, C.A., Ashwood, E.R., Bruns, D.E., Eds.; Elsevier Sounders: St. Louis, MO, USA, 2012; pp. 2131–2188. [Google Scholar]
  293. Coskun, A. Diagnosis based on population data versus personalized data: The evolving paradigm in laboratory medicine. Diagnostics 2024, 14, 2135. [Google Scholar] [CrossRef]
  294. Ozarda, Y.; Sikaris, K.; Streichert, T.; Macri, J. Distinguishing Reference Intervals and Clinical Decision Limits—A Review by the IFCC Committee on Reference Intervals and Decision Limits. Crit. Rev. Clin. Lab. Sci. 2018, 55, 420–431. [Google Scholar] [CrossRef] [PubMed]
  295. Fraser, C.G. Reference Change Values. Clin. Chem. Lab. Med. 2012, 50, 807–812. [Google Scholar] [CrossRef]
Figure 1. Biomarkers used as established tumor markers and potential tumor markers under investigation. (A) Frequently used tumor markers. Color coding: orange (protein, Section 2.1), blue (enzyme, Section 2.2), purple (hormone, Section 2.3), and green (carbohydrate antigen, Section 2.4). Created in BioRender. Coskun, A. (2025) https://BioRender.com/ow3kws3. (B) Novel tumor markers. Color coding: green (genetic alterations), brown (tissue), red (blood), orange (urine), and purple (breath). Created in BioRender. Coskun, A. (2025) https://BioRender.com/2cof13n. Except for ovarian and prostate cancer, the cancers shown are not gender-specific. The associated references for novel tumor markers are provided in the Supplementary Materials, Tables S1–S4.
Figure 1. Biomarkers used as established tumor markers and potential tumor markers under investigation. (A) Frequently used tumor markers. Color coding: orange (protein, Section 2.1), blue (enzyme, Section 2.2), purple (hormone, Section 2.3), and green (carbohydrate antigen, Section 2.4). Created in BioRender. Coskun, A. (2025) https://BioRender.com/ow3kws3. (B) Novel tumor markers. Color coding: green (genetic alterations), brown (tissue), red (blood), orange (urine), and purple (breath). Created in BioRender. Coskun, A. (2025) https://BioRender.com/2cof13n. Except for ovarian and prostate cancer, the cancers shown are not gender-specific. The associated references for novel tumor markers are provided in the Supplementary Materials, Tables S1–S4.
Biomolecules 15 01011 g001
Figure 2. Schematics of the portable biosensor for prostate cancer diagnostics. (A) General detection process and smartphone-mediated readout. (B) Electrode system and fabrication process. (C) Surface modification process and PSA detection. PLL: poly-L-lysinehydrobromide; BSA: bovine serum albumin. (Adapted with permission from Reference [205], published by Elsevier 2023, https://doi.org/10.1016/j.bios.2023.115193.
Figure 2. Schematics of the portable biosensor for prostate cancer diagnostics. (A) General detection process and smartphone-mediated readout. (B) Electrode system and fabrication process. (C) Surface modification process and PSA detection. PLL: poly-L-lysinehydrobromide; BSA: bovine serum albumin. (Adapted with permission from Reference [205], published by Elsevier 2023, https://doi.org/10.1016/j.bios.2023.115193.
Biomolecules 15 01011 g002
Figure 3. Non-invasive early detection strategies for breast cancer. (A) Microneedle patch that enables minimally invasive and rapid removal of interstitial fluid for CEA and CA 15-3 measurements (Adapted with permission from Reference [245], published by Elsevier 2023, https://doi.org/10.1016/j.cej.2023.145036). (B) A portable plasmonic SERS platform based on Raman scattering profiles of tear fluid for breast cancer detection. Au/HCP-PS: gold-decorated, hexagonal-close-packed polystyrene; PC-LDA: principal component linear discriminant analysis. (Adapted with permission from Reference [220], copyright (2020) American Chemical Society, https://doi.org/10.1021/acsami.9b19421.
Figure 3. Non-invasive early detection strategies for breast cancer. (A) Microneedle patch that enables minimally invasive and rapid removal of interstitial fluid for CEA and CA 15-3 measurements (Adapted with permission from Reference [245], published by Elsevier 2023, https://doi.org/10.1016/j.cej.2023.145036). (B) A portable plasmonic SERS platform based on Raman scattering profiles of tear fluid for breast cancer detection. Au/HCP-PS: gold-decorated, hexagonal-close-packed polystyrene; PC-LDA: principal component linear discriminant analysis. (Adapted with permission from Reference [220], copyright (2020) American Chemical Society, https://doi.org/10.1021/acsami.9b19421.
Biomolecules 15 01011 g003
Figure 4. Population-based and personalized reference intervals for tumor markers. (A) The popRI and prRI for CA 19-9 are shown as an example. Reference intervals can differ substantially between the population and the individual. As illustrated in the figure, if the individual’s measurement result is 30 kU/L, it would be considered normal when interpreted using the popRI but classified as abnormal when using the prRI. (B) Personalized reference intervals (prRIs) and population-based reference intervals (popRIs) represent different levels of tumor marker interpretation. For a given tumor marker, the popRI often does not reflect the individual’s physiological baseline, resulting in potentially low specificity when applied at the individual level. In contrast, prRIs enhance the specificity of reference intervals, allowing for more accurate distinction between healthy and pathological states in individual patients. (B): Reprinted with permission from Reference [293], published by MDPI AG 2024, https://doi.org/10.3390/diagnostics14192135.
Figure 4. Population-based and personalized reference intervals for tumor markers. (A) The popRI and prRI for CA 19-9 are shown as an example. Reference intervals can differ substantially between the population and the individual. As illustrated in the figure, if the individual’s measurement result is 30 kU/L, it would be considered normal when interpreted using the popRI but classified as abnormal when using the prRI. (B) Personalized reference intervals (prRIs) and population-based reference intervals (popRIs) represent different levels of tumor marker interpretation. For a given tumor marker, the popRI often does not reflect the individual’s physiological baseline, resulting in potentially low specificity when applied at the individual level. In contrast, prRIs enhance the specificity of reference intervals, allowing for more accurate distinction between healthy and pathological states in individual patients. (B): Reprinted with permission from Reference [293], published by MDPI AG 2024, https://doi.org/10.3390/diagnostics14192135.
Biomolecules 15 01011 g004
Figure 5. Decision limits are critical thresholds used to diagnose diseases based on biomarker levels. Currently, these limits are typically derived from population-based data. Many tumor markers exhibit marked individuality, a strong need for personalization. Since personalized reference intervals (prRIs) and population-based reference intervals (popRIs) differ significantly for the same tumor marker, the corresponding decision limits may also vary. This discrepancy highlights the need to personalize decision limits to improve diagnostic accuracy at the individual level. UL: upper limit; LL: lower limit; popDL: population-based decision limit; and PrDL: personalized decision limit.
Figure 5. Decision limits are critical thresholds used to diagnose diseases based on biomarker levels. Currently, these limits are typically derived from population-based data. Many tumor markers exhibit marked individuality, a strong need for personalization. Since personalized reference intervals (prRIs) and population-based reference intervals (popRIs) differ significantly for the same tumor marker, the corresponding decision limits may also vary. This discrepancy highlights the need to personalize decision limits to improve diagnostic accuracy at the individual level. UL: upper limit; LL: lower limit; popDL: population-based decision limit; and PrDL: personalized decision limit.
Biomolecules 15 01011 g005
Figure 6. The reference change value (RCV) indicates the magnitude of significant change between serial measurements and is a critical parameter in disease monitoring using biomarkers. Although sequential results may fall within the reference interval and be considered normal by conventional approaches, they can, in fact, differ significantly from each other. The RCV helps to identify such clinically meaningful changes, even when values remain within the population-based reference interval. The yellow and red circles represent the first and second measurement results of the same tumor markers in samples taken at different time points.
Figure 6. The reference change value (RCV) indicates the magnitude of significant change between serial measurements and is a critical parameter in disease monitoring using biomarkers. Although sequential results may fall within the reference interval and be considered normal by conventional approaches, they can, in fact, differ significantly from each other. The RCV helps to identify such clinically meaningful changes, even when values remain within the population-based reference interval. The yellow and red circles represent the first and second measurement results of the same tumor markers in samples taken at different time points.
Biomolecules 15 01011 g006
Table 1. Tumor volume doubling times in different cancers, key tumor markers, and their index of individuality.
Table 1. Tumor volume doubling times in different cancers, key tumor markers, and their index of individuality.
CancerSubtypeTumor Volume
Doubling Time
Tumor MarkerIIRef.
Lymphoid
neoplasms
Burkitt’s lymphoma24–48 h LDH0.37 *[10]
Testicular cancer Non-seminoma21 days AFP0.08 *[11]
Brain tumorGlioblastoma29.8 days VEGF-[12]
Lung cancerSmall cell lung cancer73 days * ¤NSE0.66 *[13]
proGRP0.29
Ovarian cancer 90 days CA 1250.34 *[14]
Lung cancerSquamous cell
lung cancer
140 days * ¤SCC-[13]
Liver cancerHepatocellular
carcinoma
140 days * ¤AFP0.08 *[15]
Pancreatic cancer 144 days CA 19-90.07 *[16]
Breast cancer 180 days * CA 15-30.12[17]
Gastric cancer 186 days (T1) CEA0.11 *[18]
CA 19-90.07 *
Colorectal cancer 211 days CEA0.11 *[19]
Lung cancerAdenocarcinoma223 days * ¤CEA0.11 *[13]
CYFRA 21.10.67
Thyroid cancerMedullary thyroid
carcinoma
1.6 years Calcitonin0.2 *[20]
Thyroglobulin0.14 *
Prostate cancer >2 year
(89% of patients)
PSA0.16 *[21,22]
Thyroid cancerPapillary thyroid
carcinoma
>5 year
(71.8% of patients)
Thyroglobulin0.14 *[23]
II: The index of individuality was calculated using data provided in the EFLM database. * The data presented are based on systematic review and/or meta-analysis. ¤ Pooled mean, mean, and median.
Table 2. Overview of key tumor markers: measurement methods, clinical applications, and year of investigation.
Table 2. Overview of key tumor markers: measurement methods, clinical applications, and year of investigation.
Tumor MarkersMethodPrimary
Malignancy
Other
Malignancies
Non-Malignant
Conditions
YearRef.
Bence Jones
Protein
IFE,
SFLC
Multiple
myeloma *
Non-Hodgkin’s lymphoma,
Waldenström’s macroglobulinemia
Pre-malignant plasma cell disorders 1847[27]
hCGECLIA, CLIAGerm cell and testicular tumors, gestastional trophoblastic neoplasia *Lung cancerHyperthyroidism, chronic renal failure1956[28]
AFPECLIA, CLIAHepatocellular carcinoma *, germ cell tumors *Gastric, colorectal, bilary, pancreatic, and lung cancerLiver regeneration, viral hepatitis, pregnancy1963[29]
CEAECLIA, CLIAColorectal cancerBreast, lung, gastric, pancreatic, bladder, cervical, thyroid, and hepatic cancers, lymphoma and melanoma Ulcerative pancreatitis, cirrhosis, colitis, hypothyroidism, Crohn’s disease, COPD1965[30]
NSEECLIA, TRACENeuroendocrine tumors (neuroblastoma, small cell lung cancer)Medullary thyroid carcinoma, melanoma, pancreatic
endocrine tumors
Tuberculosis, COPD, alveolar proteinosis, acute respiratory distress syndrome, silicosis, neurological deficits, ischemia reperfusion, brain injury1965[31]
Chromo-granin ATRACENeuroendocrine tumorsPresence of neuroendocrine cells in non-endocrine tumors Atrophic gastritis, chronic renal injury, chronic heart failure, hypertension, rheumatoid arthritis 1967[32]
CalcitoninECLIA, ICMAMedullary thyroid carcinoma *Lung, breast, kidney, and liver cancerPulmonary disease, pancreatitis, hyperparathyroidis, pernicious anemia1968[33]
Thyro-globulinLC-MS/MSThyroid cancerNoneGraves’ disease, Hashimoto’s disease, and thyroiditis1975[34]
SCCATRACESquamous cell carcinoma (cervical, lung,
skin, head and neck)
Esophageal adenocarcinoma, hepatocellular carcinomaInverted papilloma, non-malignant pulmonary disease, chronic hepatitis, atopic dermatitis 1977[35]
PSAECLIA, CLIAProstate cancer *NoneUrinary tract infections, prostatisis, benign prostatic hyperplasia1979[36]
CA 19-9ECLIA, CLIAPancreatic
cancer *
Colorectal, biliary tract, liver, gastric, and lung cancer, cholangiocarcinoma,
mesothelioma
Liver damage, bile duct obstruction and inflammation, pancreatitis, interstitial pulmonary disease, pulmonary fibrosis, collagen vascular diseases, hypothyroidism, gastric ulcer1979[37]
CA 125ECLIAOvarian cancer *Breast, endometrial, cervix, peritoneal, uterus, lung, and pancreatic cancer, non-Hodgkin lymphoma, hepatocellular carcinomaIdiopathic pulmonary fibrosis, ovarian cyst, endometriosis, adenomyosis, pelvic inflammation, uterine fibroids, rheumatoid arthritis-related interstitial lung disease1981[38]
CA 15-3ECLIABreast cancerPancreatic, lung, ovarian, colorectal, and liver cancerBenign liver and breast diseases1984[39]
Inhibin A
Inhibin B
ICMA
ELISA
Ovarian granulosa cell, mucinous epithelial ovarian and testicular tumorsEndometrial carcinoma, adrenal tumorsPreeclampsia, ovarian cysts1989[40]
HE4ECLIAOvarian cancerLung cancer, pulmonary adenocarcinomaChronic kidney disease, renal failure, kidney fibrosis1991[41]
Cyfra 21.1ECLIALung cancerBreast, bladder, and pancreatic cancer, hepatocellular carcinomaRenal failure, liver cirrhosis, benign lung diseases 1993[42]
COPD: chronic obstructive pulmonary disease, ECLIA: electrochemiluminescence immunoassay; CLIA: chemiluminescent immunoassay; LC-MS/MS: liquid chromatography tandem mass spectrometry; ICMA: immunochemiluminometric assay; TRACE: time-resolved amplified cryptate emission cryptate emission; IFE; immunofixation electrophoresis; and SFLC: serum free light chain. * Current recommended clinical applications involve diagnosis.
Table 3. Biosensors used in portable devices for measuring tumor markers in various body fluids.
Table 3. Biosensors used in portable devices for measuring tumor markers in various body fluids.
BiofluidBiomarkerMethodDetection
Limit
Assay
Time
Measurement ProceduresRef.
Serum CEAOptical (fluorescence quenching)6.7 pg/mL 80 minPaper-based device with mesoporous silica NP and quantum dot signal generation via glucose-triggered fluorescence quenching.[193]
CEAPhotoelectrochemical 11.3 pg/mL~35 minPaper-based immunoassay platform integrating shell–shell structured photoactive materials.[194]
CEAOptical (scanned image analysis)0.45 ng/mL15 minLateral flow strip with Au-NP probes and nitrocellulose membranes. Office-type scanner used for quantification.[195]
CEAOptical (SERS)0.36 pg/mL ~30 minPump-free microfluidic chip using a Au-NP-modified SiO2 microsphere.[196]
SCCA0.45 pg/mL
CA 19-9Electrochemical (DPV)0.07 U/mL ~25 minFlexible SP carbon electrode modified with carbon black-polyelectrolyte multilayer films.[197]
CA 19-9Optical 30 U/mL 35 minLateral flow sensor integrating magnetized CNT for low-cost, visual detection on disposable strips.[198]
CA 125Optical (colorimetric)5.21 U/mL20 minLateral flow platform utilizing Au nanozyme-labeled probes for low-cost, home-usable, and visually quantitative detection.[199]
CA 125Electrochemical (DPV) 2 mU/mL ~25 minSmartphone-integrated system combining a miniaturized detector and SP electrodes modified with CNT and Au-NP.[200]
CA 15-3Electrochemical, (SWV) 0.95 U/mL ~30 minDisposable chip device based on NP-modified SP electrodes.[201]
AFPElectrochemical, (SWV)0.03 ng/mL35 sPOC biosensor integrating multi-functionalized graphene nanocomposites for rapid biomarker detection.[202]
AFPPhotoelectrochemical 74.8 pg mL 1.5 hBiosensor combining oxygen-doped semiconductor photoelectrodes with digital multimeter readout for simple and low-cost biomarker analysis.[203]
AFPOptical1.27 ng/mL2 h Droplet evaporation-based biosensor utilizing surfactant-modified patterns on plastic substrates for simple, label-free detection.[204]
PSAElectrochemical (DPV)0.38 fg/mL20 minMiniaturized sensor integrating shrink polymer-based electrodes with smartphone-controlled operation.[205]
LDHOptical (colorimetric)70 pg mL 50 minElectrophoretic lateral flow sensor integrating battery-powered microfluidics, Au-NP signal transduction, and smartphone-based signal quantification. Small benchtop set-up needed.[206]
LDHOptical (colorimetric)86 ng/mL (LOQ)10 minSmartphone-based lateral flow biosensor using carbon NP for visual detection on disposable strips.[207]
ThyroglobulinOptical (LSPR)93.11 fg/mL10 minFiber optic localized surface plasmon resonance biosensor integrating Au-NP-coated fibers within a microfluidic channel for simplified detection.[208]
miRNA 21 Electrical (direct current)0.0028 fM 1.5 hSelf-powered platform employing graphdiyne-modified electrodes, physical signal amplification, and smartphone readout.[209]
Whole bloodCEAElectrochemical (linear sweep voltammetry)0.15 ng/mL~25 minFluidic-integrated dual carbon electrode platform fabricated by stencil printing. [210]
CA 1250.6 U/mL
CEAOptical (fluorescence)10 ng/mL~10 minMicrofluidic silk patch fabricated by 3D printing for flexible sensing.[211]
AFP10 ng/mL
PSAOptical (fluorescence) 0.08 ng/mL13–22 minPower-free and flexible, fluoropolymer
microcapillary film device integrated with a smartphone.
[212]
NSEEIS1.15 ng/mL 5 minDisposable chip-like device, enabling simplified detection using mouse model samples without clinical validation.[213]
SalivaCEAOptical (fluorescence)0.012 ng/mL~5 minFully integrated platform combining acoustic enrichment and smartphone-based visual detection for easy home monitoring.[214]
CEAOptical (time-resolved photoluminescence)1.47 pg/mL10 minLab-in-syringe platform integrating lanthanide nanoprobes with dissolution-enhanced luminescence for easy on-site detection.[215]
CYFRA 21-1Electrochemical (DPV, chronoamperometry)0.025 ng/mL (LLOQ) 4 hPaper-based platform with silver nano-ink printed electrodes.[216]
CEAElectronic (direct current measurement)0.148
pg/mL
1 h Label-free biosensor integrating rGO/melamine-modified electrodes with wired electronic readout system.[217]
CYFRA 21-10.04 pg/mL
CA 15-3Electrochemical (DPV)0.56 U/mL 1 hImmunosensor integrating SP paper electrodes modified with AuNPs for simple detection on disposable platforms.[218]
UrineNMP22, CA9, CD47, CK8, CK18 Electrical (FET)≪pg/mL~5 minIGZO FET-based urinalysis device integrated with wireless data transfer and smartphone interface for the simultaneous detection of five bladder cancer markers.[219]
TearsRaman spectral profileOptical (SERS)100 fM ~5 minLabel-free Au/HCP-PS biosensor combined with a hand-held Raman spectrometer enables the detection of breast cancer with 96% classification accuracy.[220]
Artificial sampleNSEEIS1.005 ng/mL 5 minMicrofluidic chip incorporating Au-modified electrodes for simplified detection, without clinical validation.[221]
CEA Optical (fluorescence)3.1 ng/mL 20 minMicrofluidic device combining magnetic single-bead trapping with acoustic micro-mixing. Small benchtop set-up needed.[222]
PSA0.028 ng/mL
CA 15-3Electrochemical (SWV)0.909 mU/mL 20 minDisposable sensor platform utilizing MIPs as alternative to natural sensing elements for stable and selective detection.[223]
SERS: surface-enhanced Raman scattering; SCCA: squamous cell carcinoma antigen; si: silica; DPV: differential pulse voltammetry; CNT: carbon nanotube; SWV: square wave voltammetry; POC: point of care; Au/HCP-PS: gold-decorated/hexagonal-close-packed polystyrene; LSPR: localized surface plasmon resonance, rGO: reduced graphene oxide; FET: field-effect transistor; SP: screen printed; EIS: electrochemical impedance spectroscopy.
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

Savas, I.N.; Coskun, A. The Future of Tumor Markers: Advancing Early Malignancy Detection Through Omics Technologies, Continuous Monitoring, and Personalized Reference Intervals. Biomolecules 2025, 15, 1011. https://doi.org/10.3390/biom15071011

AMA Style

Savas IN, Coskun A. The Future of Tumor Markers: Advancing Early Malignancy Detection Through Omics Technologies, Continuous Monitoring, and Personalized Reference Intervals. Biomolecules. 2025; 15(7):1011. https://doi.org/10.3390/biom15071011

Chicago/Turabian Style

Savas, Irem Nur, and Abdurrahman Coskun. 2025. "The Future of Tumor Markers: Advancing Early Malignancy Detection Through Omics Technologies, Continuous Monitoring, and Personalized Reference Intervals" Biomolecules 15, no. 7: 1011. https://doi.org/10.3390/biom15071011

APA Style

Savas, I. N., & Coskun, A. (2025). The Future of Tumor Markers: Advancing Early Malignancy Detection Through Omics Technologies, Continuous Monitoring, and Personalized Reference Intervals. Biomolecules, 15(7), 1011. https://doi.org/10.3390/biom15071011

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