Cancer Biomarkers: Comparison
Please note this is a comparison between Version 2 by Catherine Yang and Version 1 by Jagadeeswara rao Bommi.

Cancer is one of the major public health issues in the world. It has become the second leading cause of death, with approximately 75% of cancer deaths transpiring in low- or middle-income countries. It causes a heavy global economic cost estimated at more than a trillion dollars per year. The conventional diagnostic technologies such as MRI (magnetic resonance imaging), CT (computerized tomography) scan, ultrasound and biopsy were not effective for cancer detection at primary stages; this is because of their dependency on tumorigenic properties or phenotypic characters of a tumor. Cancer is a very complex disease, with many epigenetic as well as genetic modifications which might alter the cell signaling process, related to development and resulting in tumorigenic transformation and malignancy. For almost all cancer patients, researchers and clinicians expect tests or methods that might diagnose cancer significantly earlier, provide better prognosis, and that can allow for increased survival rates. Cancer markers have been used in the oncology field. Biomarkers are molecules of biologic emergence found in blood, tissues, various body fluids such as urine, cerebrospinal fluid, or different body tissues that are elevated is the indicative of an abnormal disease or condition with cancer. 

  • Cancer Biomarkers
  • Cancer Diagnosis

1. Clinical Significance of Cancer Biomarkers

Cancer biomarkers can be utilized for cancer patient evaluation in different clinical levels, as well as disease screening, prognosis, diagnosis, staging, risk assessment, stratification, therapy planning and monitoring (Figure 21). Still, to date, several cancer markers have indicated poor validity and efficacy, especially in the most widespread cancers such as lung and breast cancers. Cancer biomarkers are bio-molecules necessary for remodeling throughout cancer which maintain excessive clinical relevance. These can be enzymes, iso-enzymes, nucleic acids, proteins, metabolites. Biomarkers are classified into three types based on their clinical advantage: prognostic, predictive and diagnostic. Prognostic biomarkers provide details about course of recurrence of the disease; patient response to the treatment is estimated by predictive biomarkers; disease detection can be performed by using diagnostic biomarkers [43,47,48,49][1][2][3][4]. The difference in the level of any unique biomarker in a cell or tissue is often used as evidence of tumor expansion. Biomarkers also play potential role in differentiating benign and malignant tumors and one type of malignancy from another type; specific biomarkers are helpful in unique settings, other biomarkers can be involved in multiple settings [48][3]. Biomarkers might be helpful to estimate a person’s chance of developing tumors/cancer. For example, a person having a solid family network (via ancestors) with ovarian cancer might receive a genetic test to decide whether they are acting as a carrier for a specific germ line modification or mutation, such as BRCA1, which could cause potential chance of developing breast and/or ovarian cancer [50,51][5][6]. Biomarkers might be helpful to determine malignancy in fit populations. A frequently utilized component for screening is PSA (prostate-specific antigen). It was approved by Food and Drug Administration (FDA) in 1986. Enhanced screening in male population above 50 years old lead to growth in the identification of prostate cancer. These kinds of traditional biomarkers also have limitations. In the previous decade, U.S. Preventive Services Task Force survey noted that an adequate documentation for common diagnosis with PSA [52,53,54][7][8][9]. Biomarkers were used to monitor prognosis and possibility of cancer reappearance irrespective of therapy/treatment. The clinical and pathological properties of a tumor could be useful for the prediction of cancers. Recently, modern techniques were used to evaluate prognosis of independent tumors; for example, a large number of genetic marks that had been matured in breast cancer might be useful to evaluate the identification for an individual patient depending on tumor assessment [55,56][10][11]. During breast cancer (metastatic) condition, circulating tumor cells are indicative of overall survival [57,58,59][12][13][14]. Biomarkers could be utilized as stimulus changers or prognostic factors for unique type of therapy, as well as to select the effective type of treatment. KRAS is a predictive biomarker for colorectal cancer; mutations occurring to KRAS in somatic cells are related to low response to anti-EGFR-mediated treatment [60,61][15][16]. Likewise, HER2 over-expression in the breast cancer as well as gastric cancer anticipates for stimulus to anti-Her2 drugs such as trastuzumab [62,63,64,65[17][18][19][20][21][22],66,67], and over-expression of estrogen receptor anticipates for stimulus to anti-endocrine therapy or treatment such as tamoxifen in breast cancer [68,69][23][24]. Identification of novel cancer biomarkers might help in quick and efficient diagnosis as well as monitoring of cancer progression.
Figure 21.
Cancer biomarker clinical applications.

2. Identification of Novel Cancer Biomarkers

Possible cancer biomarkers could be recognized through various approaches. An excellent way to recognize novel biomarkers mostly depends on biological nature of tumor and nearby environment of a tumor or metabolism of the drugs or biological products. With most recent studies and new information related to cancers and appearance of latest technology, cancer biomarker detection is performed frequently these days with applying a discovery approach. In this approach, few major areas of research such as gene expression arrays, proteomic technologies (mass spectroscopy, LC-MS/MS, MALDI-MS), and high throughput sequencing can be used to rapidly recognize unique biomarkers or pool of biomarkers which can show difference in the middle of cohorts. Expansion of sophisticated software algorithms for large data analysis has emerged in rapid advancement in the identification of novel cancer biomarkers. Openly available software programs can sort these data and compare the sequences to annotated genome databases to permit quantitative comparative evaluation of proteomes from multiple sources such as tumor area and nearby healthy tissues. Thus, over-expressed or down-regulated proteins in a cancer cell can be identified as putative cancer biomarkers. Huge amount of data produced with these technological methods mean that special attention needs to be directed toward both developing a study plan and conducting a large data analysis. Moreover, it is crucial to reduce the possibility of detecting relationships that are eventually determined to provide false-positive results. The most crucial features of biomarker improvement and identification to consider in depth include mindful study pattern to minimize any kind of bias, extensive evaluation, validation, and accurate communication of the results [48,70,71,72,73,74,75,76,77,78,79][3][25][26][27][28][29][30][31][32][33][34].

3. Cancer Biomarkers Currently Used in Clinical Settings

A cancer biomarker is a molecule existing inside and/or generated by tumor cells or surrounding cells in tissues or organs in stimulus to tumor or certain noncancerous (benign) situations, which can provide information about cancer, mostly which stage it is, whether it can respond to treatment, and what type of therapy might be useful. Here, few recent cancer biomarkers used in clinical practice are explained. New cancer biomarkers become available continuously, and they may not be explained below [27,40,44,80,81,82][35][36][37][38][39][40].

3.1. AFP (Alpha-Fetoprotein)

AFP (Alpha-fetoprotein) is one of the leading biomarker. Early fetal life of a baby (mostly yolk sac and liver) produces AFP during pregnancy. AFP can be detected in huge amounts in serum of the patients with specific tumors. According to Yuri Semenovich Tatarinov, AFP was first accepted as an antigen unique for human HCC (hepatocellular carcinoma) [83,84][41][42]. The scientific literature has explained that AFPs are classified into subtypes based on their dissimilar affinities to LCA (lens culinaris agglutinin), such as AFP-L1 (LCA unreactive AFP), AFP-L2 (LCA mild active AFP) and AFP-L3 (LCA active AFP). In healthy individual serum, an average level of AFP is less than 20 ng 8 mL−1 [85,86,87][43][44][45]. AFP is widely established for HCC (hepatocellular carcinoma) diagnosis. Moreover, in congenital tyrosinemia, cirrhosis, hepatitis (alcohol-induced), hepatitis (viral-induced), ataxia-telangiectasia syndrome or in several malignancies such as testicular cancer, liver cancer, gastric cancer and nasopharyngeal cancer elevated AFP levels may also be present. Hence, sensing the AFP values is completely mandatory in clinical settings. High recognition rates of molecular assays have been obtained in quantitative observation of AFP due to their specificity and unalterable affinity of the probes to molecular targets [88,89][46][47].

3.2. PSA (Prostate Specific Antigen)

PSA (prostate-specific antigen) was one of the first recognized cancer biomarkers, utilized to detect and screen prostate cancer in clinical setting. It has been shown that increased levels of PSA can directly relate to prostate cancer. Human regular PSA level is 4 ng/mL. According to the study by Smith et. al, almost 30% of individuals with PSA values higher than normal (range of 4.1–9.9 ng/mL) were diagnosed with prostate cancer [90][48]. Along with this, raised PSA values may also indicate benign tumors (non-fatal), prostatitis/prostate inflammation or benign prostatic hyperplasia. Therefore, elevated values of PSA may not consistently suggest malignant tumors. There is a fact that caused reasonable controversy about using regular PSA screening to detect prostate cancer. Small-sized tumors identified by PSA screening may grow very slowly; death caused by small tumor might not be feasible in an individual lifetime. Moreover, it is very expensive to treat such slow-growing tumors. Other frequent issue with PSA screening is false-positives. This limitation of PSA testing can be overcome by modern biosensing technology mediated by biosensors [79,84,91,92,93][34][42][49][50][51]. PSA detection can be performed by various methods; these traditional methods are time-consuming as well as expensive. Yang et al. explained a graphene oxide/ssDNA-based biosensor integrated with dual antibody-modified PLLA NPs to amplify electrochemical signals for the effective and rapid electrochemical capture of PSA in serum samples from prostate cancer patients. The detection limit for PSA was 1 ng/mL, which achieved a wide linear range of 1–100 ng/mL for PSA. This is one of the examples that shows the usefulness of modern biosensing technology mediated by biosensors [94][52].

3.3. RCAS1 (Receptor-Binding Cancer Antigen)

RCAS1 (receptor-binding cancer antigen) overexpression data has been described in many gastric carcinomas; it is related with progression of gastric cancer. Further, RCAS1 is also suggested as a cancer biomarker for poor prediction in breast, esophageal, endometrial, bladder cancers and is associated with tumor weakening in pharyngeal carcinoma and laryngeal cancer. RCAS overexpression is observed in several types of cancers. Thus, it serves as a potential biomarker for cancer detection and prediction [95,96,97,98][53][54][55][56].

3.4. CA 15-3 (Cancer Antigen 15-3)

The most predominant cancer marker for breast cancer identification as well as monitoring is cancer antigen 15-3; additional biomarkers that are related to breast cancer are CA 27.29, BRCA1, BRCA2 and (carcinoembryonic antigen) CEA [44,99,100][37][57][58]. This particular marker is frequently used on a clinical level to monitor the therapy for breast cancer in its advanced stages. During breast cancer, CA 15-3 values increased by 10, 20, 30 and 40 % at various stages first, second, third and fourth stage [49][4]. Tampellini et. al showed the connection between breast cancer and CA 15-3 levels, also mentioning that before treatment, patients with levels of 30U/mL had notably higher survival rate compared to patients with higher values. Raised CA 15-3 values correspond with extensive metastasis [101,102][59][60]. Other research data explained that raised CA 15-3 levels post cancer treatment can be a sign of disease recurrence. Nowadays, to determine breast cancer treatment, protocol CA 15-3 values are considered along with risk factors (negative) such as PR/ER condition as well as iHer-2, cancer stage and tumor dimension. Hepatitis, endometriosis, pelvic inflammatory disease, lactation and pregnancy are conditions other than cancer where CA 15-3 levels are increased [103][61].

3.5. Cancer–Testis (CT) Antigens

Cancer–testis antigens are a specific type of cancer biomarkers. They are expressed in various cancers. CT antigen expression is limited to male germ cells of the testis but not shown in normal adult cells. These antigens are also expressed in trophoblast and ovary cells. Therefore, CT antigens have been considered as possible immunogenic targets for cancer vaccines (cancer immunotherapies). CT antigen autoantibodies have been studied as potential cancer biomarkers [104][62]. NY-ESO-1 (NewYorkesophagealsquamouscellcarcinoma1) is encoded by CTAG-1B; this is the class of antigens with high immunogenic nature which induces very robust cellular and humoral immune response in NY-ESO-1-positive cancers. Antibody titer to NY-ESO-1 has been shown to relate with disease progression. One of the best benefits of using CT autoantibodies as tumor biomarkers is the fact that they are easy to obtain and are also more stable proteins present in serum compared to tissues obtained via biopsy. Thus, they can be useful for cancer progression and recurrence [105,106,107,108,109][63][64][65][66][67]. Major limitation of CT antigens is the fact that many cancers express CT antigens, and they are rarely tumor-specific. Biosensor technology can obtain a profile for CT antigen, which can enhance the use of these antigens in cancer prognosis and diagnosis [110,111,112][68][69][70].

3.6. CA 125

The rise in CA 125 levels is primarily related to ovarian cancer. It is further correlated with several different cancers such as cervix, lungs, breast, liver, pancreas, uterus, stomach and colon cancers. Enhanced levels of CA 125 are also observed in various non-pathological conditions such as menstruation and pregnancy [8,87,113][45][71][72]. A total of 90% of women with advanced stage ovarian cancer and 40% of humans with intra-abdominal malignant tumors also exhibit high CA 125 levels. Still, approximately 50% of patients diagnosed with primary stage ovarian cancer show normal CA 125 levels [87,88,89][45][46][47]. Other germ cell origin biomarkers such as alpha-fetoprotein/AFP, human chorionic gonadotrophin/HCG as well as Lactate dehydrogenase/LDH are also connected to ovarian cancer [114,115][73][74]. Increase in CA 125 levels can be used to detect the development of benign tumors into malignant tumors. Enhanced CA 125 levels are also used to identify treatment failure as well as disease recurrence (e.g., high CA 125 levels after bilateral salpingo-oophorectomy or total abdominal hysterectomy, which may occur after first line chemotherapy) [116,117,118,119,120][75][76][77][78][79]. Altogether, CA 125 is an extremely essential biomarker for detection of cancer, and also for cancer progression monitoring and treatment.

3.7. CA 19-9

This antigen was first identified in pancreatic and colon cancer patient’s serum in 1981. It is a Lewis antigen of the MUC1 glycoprotein [8,121][71][80]. CA 19-9 normal level in serum is less than 37 U/mL. In the recent decade, on a clinical level, CA 19-9 biomarker has become extremely useful for the diagnosis of pancreatic cancer. Normal human serum CA 19-9 levels can play an outstanding role in clinical diagnosis of urothelial and gastric cancers [122,123][81][82]. Consequently, there is a necessity to improve highly sensitive methods which can detect CA 19-9 values in patients with cancer.

3.8. Nse (Neuron-Specific Enolase)

This neuron-specific enolase is a popular and unique marker for SCLC as well as NSCLC non-small cell lung cancer. It has a crucial role in glycolysis; in 1980s, NSE expression was noted in SCLC cells. From that time, it has been used as potential biomarker for lung cancer, able to detect increased values of NSCLC and acting as crucial predictor for patient survival (it is not dependent on remaining prognostic factors) [124,125,126][83][84][85]. NSE is also a unique marker for neuro-endocrine cells. Raised NSE values in body fluids might be an indication of tumor proliferation and staging determination in some of brain tumors [127,128][86][87]. In the recent decade, the value of NSE for prognosis in cancer patients is debatable. Therefore, it is mandatory to improve sensitive techniques to perceive Nse values in patients with cancer.

3.9. Tdt (Terminal Deoxynucleotidyl Transferase)

Tdt is an intracellular marker which has detected in the bone marrow as well as blood (mononucleate) cells in leukemia patients during diagnosis. Overall, TdT values are remarkably raised in several lymphocytic lymphomas. Tdt might be helpful to identify specific leukemia type and supportive sign for the solution of therapy [129,130,131,132][88][89][90][91].

3.10. CYFRA21-1

Cytokeratin-19 fragments/CYFRA21-1 have been extensively studied in patients with NSCLC and are widely utilized as predictive, prognostic markers. They have 56% sensitivity as well as 88% specificity when the value is >1.5 ng/mL. Researchers used maximum cut off value for CYFRA21-1 similar to ≥3 ng/mL; it had shown increased specificity at 97%. It has potential capability in lung, esophagal cancer prediction. Raised values are certain but barely sensitive. These values are strongly connected with cancer metastasis. Recent reports explained that CYFRA21-1 is used as independent prediction factor for various phases of lung cancer. This might function as definitive distinction between benign and malignant lung cancer along with clinical information [133,134,135,136,137][92][93][94][95][96].

References

  1. Jayanthi, V.S.A.; Das, A.B.; Saxena, U. Recent Advances in Biosensor Development for the Detection of Cancer Biomarkers. Biosens. Bioelectron. 2017, 91, 15–23.
  2. Chowdhury, S.G.; Ray, R.; Karmakar, P. Exosomal miRNAs—a diagnostic biomarker acting as a guiding light in the diagnosis of prostate cancer. Funct. Integr. Genom. 2023, 23, 23.
  3. Henry, N.L.; Hayes, D.F. Cancer Biomarkers. Mol. Oncol. 2012, 6, 140–146.
  4. Somerfield, M.R. Clinical Practice Guidelines for the Use of Tumor Markers in Breast and Colorectal Cancer. Adopted on May 17, 1996 by the American Society of Clinical Oncology. J. Clin. Oncol. 1996, 14, 2843–2877.
  5. Meindl, A. Comprehensive Analysis of 989 Patients with Breast or Ovarian Cancer Provides BRCA1 and BRCA2 Mutation Profiles and Frequencies for the German Population. Int. J. Cancer 2002, 97, 472–480.
  6. Easton, D.F.; Ford, D.; Bishop, D.T.; Haites, N.; Milner, B.; Allan, L.; Easton, D.F.; Ponder, B.A.J.; Peto, J.; Smith, S.; et al. Breast and Ovarian Cancer Incidence in BRCA1-Mutation Carriers. Breast Cancer Linkage Consortium. Am. J. Hum. Genet. 1995, 56, 265.
  7. Grossman, D.C.; Curry, S.J.; Owens, D.K.; Bibbins-Domingo, K.; Caughey, A.B.; Davidson, K.W.; Doubeni, C.A.; Ebell, M.; Epling, J.W.; Kemper, A.R.; et al. Screening for Prostate Cancer USPreventive Services task force recommendation Statement. JAMA-J. Am. Med. Assoc. 2018, 319, 1901–1913.
  8. Lin, K.; Lipsitz, R.; Miller, T.; Janakiraman, S. Benefits and Harms of Prostate-Specific Antigen Screening for Prostate Cancer: An Evidence Update for the U.S. Preventive Services Task Force. Ann. Intern. Med. 2008, 149, 192–199.
  9. Calonge, N.; Petitti, D.B.; Dewitt, T.G.; Dietrich, A.J.; Gregory, K.D.; Harris, R.; Isham, G.J.; Lefevre, M.L.; Leipzig, R.; Loveland-Cherry, C.; et al. Screening for Prostate Cancer: U.S. Preventive Services Task Force Recommendation Statement. Ann. Intern. Med. 2008, 149, 185–191.
  10. Van’t Veer, L.J.; Paik, S.; Hayes, D.F. Gene Expression Profiling of Breast Cancer: A New Tumor Marker. J. Clin. Oncol. Off. J. Am. Soc. Clin. Oncol. 2005, 23, 1631–1635.
  11. Paik, S.; Shak, S.; Tang, G.; Kim, C.; Baker, J.; Cronin, M.; Baehner, F.L.; Walker, M.G.; Watson, D.; Park, T.; et al. A Multigene Assay to Predict Recurrence of Tamoxifen-Treated, Node-Negative Breast Cancer. N. Engl. J. Med. 2004, 351, 2817–2826.
  12. Cohen, J. Detection and Localization of Surgically Resectable Cancers with a Multi-Analyte Blood Test. Science 2018, 359, 926–930.
  13. Cristofanilli, M.; Budd, G.T.; Ellis, M.J.; Stopeck, A.; Matera, J.; Miller, M.C.; Reuben, J.M.; Doyle, G.V.; Allard, W.J.; Terstappen, L.W.M.M.; et al. Circulating Tumor Cells, Disease Progression, and Survival in Metastatic Breast Cancer. N. Engl. J. Med. 2004, 351, 781–791.
  14. Zhao, Q.; Li, B.; Gao, Q.; Luo, Y.; Ming, L. Prognostic Value of Epithelial-Mesenchymal Transition Circulating Tumor Cells in Female Breast Cancer: A Meta-Analysis. Front. Oncol. 2022, 12, 1024783.
  15. Abe, S.; Kawai, K.; Nozawa, H.; Sasaki, K.; Murono, K.; Emoto, S.; Ozawa, T.; Yokoyama, Y.; Nagai, Y.; Anzai, H.; et al. Clinical Impact of Primary Tumor Sidedness and Sex on Unresectable Post-Recurrence Survival in Resected Pathological Stage II-III Colorectal Cancers: A Nationwide Multicenter Retrospective Study. BMC Cancer 2022, 22, 1–10.
  16. Allegra, C.J.; Jessup, J.M.; Somerfield, M.R.; Hamilton, S.R.; Hammond, E.H.; Hayes, D.F.; McAllister, P.K.; Morton, R.F.; Schilsky, R.L. American Society of Clinical Oncology Provisional Clinical Opinion: Testing for KRAS Gene Mutations in Patients with Metastatic Colorectal Carcinoma to Predict Response to Anti-Epidermal Growth Factor Receptor Monoclonal Antibody Therapy. J. Clin. Oncol. Off. J. Am. Soc. Clin. Oncol. 2009, 27, 2091–2096.
  17. Thuss-Patience, P.C.; Shah, M.A.; Ohtsu, A.; Van Cutsem, E.; Ajani, J.A.; Castro, H.; Mansoor, W.; Chung, H.C.; Bodoky, G.; Shitara, K.; et al. Trastuzumab Emtansine versus Taxane Use for Previously Treated HER2-Positive Locally Advanced or Metastatic Gastric or Gastro-Oesophageal Junction Adenocarcinoma (GATSBY): An International Randomised, Open-Label, Adaptive, Phase 2/3 Study. Lancet Oncol. 2017, 18, 640–653.
  18. Bang, Y.J.; Van Cutsem, E.; Feyereislova, A.; Chung, H.C.; Shen, L.; Sawaki, A.; Lordick, F.; Ohtsu, A.; Omuro, Y.; Satoh, T.; et al. Trastuzumab in Combination with Chemotherapy versus Chemotherapy Alone for Treatment of HER2-Positive Advanced Gastric or Gastro-Oesophageal Junction Cancer (ToGA): A Phase 3, Open-Label, Randomised Controlled Trial. Lancet 2010, 376, 687–697.
  19. Sawaki, M.; Taira, N.; Uemura, Y.; Saito, T.; Baba, S.; Kobayashi, K.; Kawashima, H.; Tsuneizumi, M.; Sagawa, N.; Bando, H.; et al. Adjuvant Trastuzumab without Chemotherapy for Treating Early HER2-Positive Breast Cancer in Older Patients: A Propensity Score-Adjusted Analysis of a Prospective Cohort Study. Breast 2022, 66, 245–254.
  20. Piccart-Gebhart, M.J.; Procter, M.; Leyland-Jones, B.; Goldhirsch, A.; Untch, M.; Smith, I.; Gianni, L.; Baselga, J.; Bell, R.; Jackisch, C.; et al. Trastuzumab after Adjuvant Chemotherapy in HER2-Positive Breast Cancer. N. Engl. J. Med. 2005, 353, 1659–1672.
  21. Perez, E.A.; Ballman, K.V.; Tenner, K.S.; Thompson, E.A.; Badve, S.S.; Bailey, H.; Baehner, F.L. Association of Stromal Tumor-Infiltrating Lymphocytes with Recurrence-Free Survival in the N9831 Adjuvant Trial in Patients with Early-Stage HER2-Positive Breast Cancer. JAMA Oncol. 2016, 2, 56–64.
  22. Romond, E.H.; Perez, E.A.; Bryant, J.; Suman, V.J.; Geyer, C.E.; Davidson, N.E.; Tan-Chiu, E.; Martino, S.; Paik, S.; Kaufman, P.A.; et al. Trastuzumab plus Adjuvant Chemotherapy for Operable HER2-Positive Breast Cancer. N. Engl. J. Med. 2005, 353, 1673–1684.
  23. Liu, J.; Gan, M.; Lin, Z.; Deng, Q.; Deng, J.; Zeng, B.; Shi, Y.; Ming, J. Clinical Features and Prognosis Analysis of Hormone Receptor-Positive, HER2-Negative Breast Cancer with Differential Expression Levels of Estrogen and Progesterone Receptors: A 10-Year Retrospective Study. Breast J. 2022, 2022, 5469163.
  24. Abe, O.; Abe, R.; Enomoto, K.; Kikuchi, K.; Koyama, H.; Masuda, H.; Nomura, Y.; Ohashi, Y.; Sakai, K.; Sugimachi, K.; et al. Relevance of Breast Cancer Hormone Receptors and Other Factors to the Efficacy of Adjuvant Tamoxifen: Patient-Level Meta-Analysis of Randomised Trials. Lancet 2011, 378, 771–784.
  25. Teutsch, S.M.; Bradley, L.A.; Palomaki, G.E.; Haddow, J.E.; Piper, M.; Calonge, N.; Dotson, W.D.; Douglas, M.P.; Berg, A.O. The Evaluation of Genomic Applications in Practice and Prevention (EGAPP) Initiative: Methods of the EGAPP Working Group. Genet. Med. Off. J. Am. Coll. Med. Genet. 2009, 11, 3–14.
  26. Hanash, S.M.; Pitteri, S.J.; Faca, V.M. Mining the Plasma Proteome for Cancer Biomarkers. Nature 2008, 452, 571–579.
  27. Mighton, C.; Shickh, S.; Aguda, V.; Krishnapillai, S.; Adi-Wauran, E.; Bombard, Y. From the Patient to the Population: Use of Genomics for Population Screening. Front. Genet. 2022, 13, 893832.
  28. Kohsaka, S.; Tada, Y.; Ando, M.; Nakaguro, M.; Shirai, Y.; Ueno, T.; Kojima, S.; Hirai, H.; Saigusa, N.; Kano, S.; et al. Identification of Novel Prognostic and Predictive Biomarkers in Salivary Duct Carcinoma via Comprehensive Molecular Profiling. Npj Precis. Oncol. 2022, 6, 82.
  29. Wang, N.; Zhang, H.; Li, D.; Jiang, C.; Zhao, H.; Teng, Y. Identification of Novel Biomarkers in Breast Cancer via Integrated Bioinformatics Analysis and Experimental Validation. Bioengineered 2021, 12, 12431.
  30. Wu, M.; Li, Q.; Wang, H. Identification of Novel Biomarkers Associated with the Prognosis and Potential Pathogenesis of Breast Cancer via Integrated Bioinformatics Analysis. Technol. Cancer Res. Treat. 2021, 20, 1533033821992081.
  31. Li, C.-J.; Lin, L.-T.; Chu, P.-Y.; Chiang, A.-J.; Tsai, H.-W.; Chiu, Y.-H.; Huang, M.-S.; Wen, Z.-H.; Tsui, K.-H.; Li, C.-J.; et al. Identification of Novel Biomarkers and Candidate Drug in Ovarian Cancer. J. Pers. Med. 2021, 11, 316.
  32. Islam, S.U.; Ahmed, M.B.; Ahsan, H.; Lee, Y.S. Role of Biomarkers in Personalized Medicine. In Cancer Biomarkers in Diagnosis and Therapeutics; Springer: Singapore, 2022; pp. 249–275.
  33. Janjua, K.A.; Shahzad, R.; Shehzad, A. Development of Novel Cancer Biomarkers for Diagnosis and Prognosis. In Cancer Biomarkers in Diagnosis and Therapeutics; Springer: Singapore, 2022; pp. 277–343.
  34. Bohunicky, B.; Mousa, S.A. Biosensors: The New Wave in Cancer Diagnosis. Nanotechnol. Sci. Appl. 2011, 4, 1–10.
  35. Nikanjam, M.; Kato, S.; Kurzrock, R. Liquid Biopsy: Current Technology and Clinical Applications. J. Hematol. Oncol. 2022, 15, 1–14.
  36. Altintas, Z.; Tothill, I. Biomarkers and Biosensors for the Early Diagnosis of Lung Cancer. Sens. Actuators B Chem. 2013, 188, 988–998.
  37. Tothill, I.E. Biosensors for Cancer Markers Diagnosis. Semin. Cell Dev. Biol. 2009, 20, 55–62.
  38. Prabhakar, B.; Shende, P.; Augustine, S. Current Trends and Emerging Diagnostic Techniques for Lung Cancer. Biomed. Pharmacother. 2018, 106, 1586–1599.
  39. Chen, M.; Wu, D.; Tu, S.; Yang, C.; Chen, D.J.; Xu, Y. A Novel Biosensor for the Ultrasensitive Detection of the LncRNA Biomarker MALAT1 in Non-Small Cell Lung Cancer. Sci. Rep. 2021, 11, 3666.
  40. Reddy, K.K.; Bandal, H.; Satyanarayana, M.; Goud, K.Y.; Gobi, K.V.; Jayaramudu, T.; Amalraj, J.; Kim, H. Recent Trends in Electrochemical Sensors for Vital Biomedical Markers Using Hybrid Nanostructured Materials. Adv. Sci. 2020, 7, 1902980.
  41. Ferraro, S.; Bussetti, M.; Bassani, N.; Rossi, R.S.; Incarbone, G.P.; Bianchi, F.; Maggioni, M.; Runza, L.; Ceriotti, F.; Panteghini, M. Definition of Outcome-Based Prostate-Specific Antigen (PSA) Thresholds for Advanced Prostate Cancer Risk Prediction. Cancers 2021, 13, 3381.
  42. Thompson, I.M.; Ankerst, D.P. Prostate-Specific Antigen in the Early Detection of Prostate Cancer. CMAJ Can. Med. Assoc. J. 2007, 176, 1853–1858.
  43. Wu, M.; Liu, H.; Liu, Z.; Liu, C.; Zhang, A.; Li, N. Analysis of Serum Alpha-Fetoprotein (AFP) and AFP-L3 Levels by Protein Microarray. J. Int. Med. Res. 2018, 46, 4297–4305.
  44. Yuan, Q.; Song, J.; Yang, W.; Wang, H.; Huo, Q.; Yang, J.; Yu, X.; Liu, Y.; Xu, C.; Bao, H. The Effect of CA125 on Metastasis of Ovarian Cancer: Old Marker New Function. Oncotarget 2017, 8, 50015–50022.
  45. Meyer, T.; Rustin, G.J. Role of Tumour Markers in Monitoring Epithelial Ovarian Cancer. Br. J. Cancer 2000, 82, 1535–1538.
  46. Chianca, M.; Panichella, G.; Fabiani, I.; Giannoni, A.; L’Abbate, S.; Aimo, A.; Del Franco, A.; Vergaro, G.; Grigoratos, C.; Castiglione, V.; et al. Bidirectional Relationship Between Cancer and Heart Failure: Insights on Circulating Biomarkers. Front. Cardiovasc. Med. 2022, 9, 936654.
  47. Tuxen, M.K.; Sölétormos, G.; Dombernowsky, P. Tumor Markers in the Management of Patients with Ovarian Cancer. Cancer Treat. Rev. 1995, 21, 215–245.
  48. Smith, D.S.; Humphrey, P.A.; Catalona, W.J. The Early Detection of Prostate Carcinoma with Prostate Specific Antigen: The Washington University Experience. Cancer 1997, 80, 1852–1856.
  49. Bill-Axelson, A.; Holmberg, L.; Ruutu, M.; Häggman, M.; Andersson, S.-O.; Bratell, S.; Spångberg, A.; Busch, C.; Nordling, S.; Garmo, H.; et al. Radical Prostatectomy versus Watchful Waiting in Early Prostate Cancer. N. Engl. J. Med. 2005, 352, 1977–1984.
  50. Jaffee, E.M.; Van Dang, C.; Agus, D.B.; Alexander, B.M.; Anderson, K.C.; Ashworth, A.; Barker, A.D.; Bastani, R.; Bhatia, S.; Bluestone, J.A.; et al. Future Cancer Research Priorities in the USA: A Lancet Oncology Commission. Lancet Oncol. 2017, 18, e653–e706.
  51. National Cancer Institute. Prostate-Specific Antigen (PSA) Test—NCI; National Cancer Institute: Bethesda, MD, USA, 2022.
  52. Kim, J.H.; Suh, Y.J.; Park, D.; Yim, H.; Kim, H.; Kim, H.J.; Yoon, D.S.; Hwang, K.S. Technological Advances in Electrochemical Biosensors for the Detection of Disease Biomarkers. Biomed. Eng. Lett. 2021, 11, 309–334.
  53. Dutsch-Wicherek, M.; Tomaszewska, R.; Lazar, A.; Wicherek, L.; Skladzien, J. The Association between RCAS1 Expression in Laryngeal and Pharyngeal Cancer and Its Healthy Stroma with Cancer Relapse. BMC Cancer 2009, 9, 35.
  54. Kazmierczak, W.; Lazar, A.; Tomaszewska, R.; Popiela, T.J.; Koper, K.; Wicherek, L.; Dutsch-Wicherek, M. Analysis of the Intensity of Immune Cell Infiltration and Immunoreactivity of RCAS1 in Diffuse Large B-Cell Lymphoma of the Palatine Tonsil and Its Microenvironment. Cell Tissue Res. 2015, 361, 823–831.
  55. Kubokawa, M.; Nakashima, M.; Yao, T.; Ito, K.I.; Harada, N.; Nawata, H.; Watanabe, T. Aberrant Intracellular Localization of RCAS1 Is Associated with Tumor Progression of Gastric Cancer. Int. J. Oncol. 2001, 19, 695–700.
  56. Naito, Y.; Okabe, Y.; Nagayama, M.; Nishinakagawa, T.; Taira, T.; Kawahara, A.; Hattori, S.; MacHida, K.; Ishida, Y.; Kaji, R.; et al. Accuracy of Differential Diagnosis for Pancreatic Cancer Is Improved in the Combination of RCAS1 and CEA Measurements and Cytology in Pancreatic Juice. Med. Mol. Morphol. 2011, 44, 86–92.
  57. Duffy, M.J. Serum Tumor Markers in Breast Cancer: Are They of Clinical Value? Clin. Chem. 2006, 52, 345–351.
  58. Hasan, D. Diagnostic Impact of CEA and CA 15-3 on Chemotherapy Monitoring of Breast Cancer Patients. J. Circ. Biomark. 2022, 11, 57–63.
  59. Tampellini, M.; Berruti, A.; Gerbino, A.; Buniva, T.; Torta, M.; Gorzegno, G.; Faggiuolo, R.; Cannone, R.; Farris, A.; Destefanis, M.; et al. Relationship between CA 15-3 Serum Levels and Disease Extent in Predicting Overall Survival of Breast Cancer Patients with Newly Diagnosed Metastatic Disease. Br. J. Cancer 1997, 75, 698–702.
  60. Lian, M.; Zhang, C.; Zhang, D.; Chen, P.; Yang, H.; Yang, Y.; Chen, S.; Hong, G. The Association of Five Preoperative Serum Tumor Markers and Pathological Features in Patients with Breast Cancer. J. Clin. Lab. Anal. 2019, 33, e22875.
  61. Bast, J.; Ravdin, P.; Hayes, D.F.; Bates, S.; Fritsche, H.J.; Jessup, J.M.; Kemeny, N.; Locker, G.Y.; Mennel, R.G.; Somerfield, M.R. 2000 Update of Recommendations for the Use of Tumor Markers in Breast and Colorectal Cancer: Clinical Practice Guidelines of the American Society of Clinical Oncology. J. Clin. Oncol. Off. J. Am. Soc. Clin. Oncol. 2001, 19, 1865–1878.
  62. Lin, M.J.; Svensson-Arvelund, J.; Lubitz, G.S.; Marabelle, A.; Melero, I.; Brown, B.D.; Brody, J.D. Cancer Vaccines: The next Immunotherapy Frontier. Nat. Cancer 2022, 3, 911–926.
  63. Chow, S.; Berek, J.S.; Dorigo, O. Development of Therapeutic Vaccines for Ovarian Cancer. Vaccines 2020, 8, 657.
  64. Jäger, E.; Chen, Y.T.; Drijfhout, J.W.; Karbach, J.; Ringhoffer, M.; Jäger, D.; Arand, M.; Wada, H.; Noguchi, Y.; Stockert, E.; et al. Simultaneous Humoral and Cellular Immune Response against Cancer-Testis Antigen NY-ESO-1: Definition of Human Histocompatibility Leukocyte Antigen (HLA)-A2-Binding Peptide Epitopes. J. Exp. Med. 1998, 187, 265–270.
  65. Tsuji, K.; Hamada, T.; Uenaka, A.; Wada, H.; Sato, E.; Isobe, M.; Asagoe, K.; Yamasaki, O.; Shiku, H.; Ritter, G.; et al. Induction of Immune Response against NY-ESO-1 by CHP-NY-ESO-1 Vaccination and Immune Regulation in a Melanoma Patient. Cancer Immunol. Immunother. 2008, 57, 1429–1437.
  66. Jäger, E.; Stockert, E.; Zidianakis, Z.; Chen, Y.T.; Karbach, J.; Jäger, D.; Arand, M.; Ritter, G.; Old, L.J.; Knuth, A. Humoral Immune Responses of Cancer Patients against “Cancer-Testis” Antigen NY-ESO-1: Correlation with Clinical Events. Int. J. Cancer 1999, 84, 506–510.
  67. Karbach, J.; Neumann, A.; Atmaca, A.; Wahle, C.; Brand, K.; Von Boehmer, L.; Knuth, A.; Bender, A.; Ritter, G.; Old, L.J.; et al. Efficient in Vivo Priming by Vaccination with Recombinant NY-ESO-1 Protein and CpG in Antigen Naive Prostate Cancer Patients. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2011, 17, 861–870.
  68. Stockert, E.; Jäger, E.; Chen, Y.T.; Scanlan, M.J.; Gout, I.; Karbach, J.; Arand, M.; Knuth, A.; Old, L.J. A Survey of the Humoral Immune Response of Cancer Patients to a Panel of Human Tumor Antigens. J. Exp. Med. 1998, 187, 1349–1354.
  69. Imai, N.; Ikeda, H.; Shiku, H. . Nihon Rinsho. Jpn. J. Clin. Med. 2012, 70, 2125–2129.
  70. Raza, A.; Merhi, M.; Inchakalody, V.P.; Krishnankutty, R.; Relecom, A.; Uddin, S.; Dermime, S. Unleashing the Immune Response to NY-ESO-1 Cancer Testis Antigen as a Potential Target for Cancer Immunotherapy. J. Transl. Med. J. Transl. Med. 2020, 18, 1–11.
  71. Patel, J.; Patel, P. Biosensors and Biomarkers: Promising Tools for Cancer Diagnosis. Int. J. Biosens. Bioelectron. 2017, 3, 00072.
  72. Babaier, A.; Mal, H.; Alselwi, W.; Ghatage, P. Low-Grade Serous Carcinoma of the Ovary: The Current Status. Diagnostics 2022, 12, 458.
  73. Neesham, D. Ovarian Cancer Screening. Aust. Fam. Physician 2007, 36, 126–128.
  74. Menon, U.; Gentry-Maharaj, A.; Hallett, R.; Ryan, A.; Burnell, M.; Sharma, A.; Lewis, S.; Davies, S.; Philpott, S.; Lopes, A.; et al. Sensitivity and Specificity of Multimodal and Ultrasound Screening for Ovarian Cancer, and Stage Distribution of Detected Cancers: Results of the Prevalence Screen of the UK Collaborative Trial of Ovarian Cancer Screening (UKCTOCS). Lancet Oncol. 2009, 10, 327–340.
  75. Wong, N.K.; Easton, R.L.; Panico, M.; Sutton-Smith, M.; Morrison, J.C.; Lattanzio, F.A.; Morris, H.R.; Clark, G.F.; Dell, A.; Patankar, M.S. Characterization of the Oligosaccharides Associated with the Human Ovarian Tumor Marker CA125. J. Biol. Chem. 2003, 278, 28619–28634.
  76. White, B.; Patterson, M.; Karnwal, S.; Brooks, C.L. Crystal Structure of a Human MUC16 SEA Domain Reveals Insight into the Nature of the CA125 Tumor Marker. Proteins Struct. Funct. Bioinform. 2022, 90, 1210–1218.
  77. Wang, Y.-S.; Ren, S.-F.; Jiang, W.; Lu, J.-Q.; Zhang, X.-Y.; Li, X.-P.; Cao, R.; Xu, C.-J. CA125-Tn ELISA Assay Improves Specificity of Pre-Operative Diagnosis of Ovarian Cancer among Patients with Elevated Serum CA125 Levels. Ann. Transl. Med. 2021, 9, 788.
  78. Seelenmeyer, C.; Wegehingel, S.; Lechner, J.; Nickel, W. The Cancer Antigen CA125 Represents a Novel Counter Receptor for Galectin-1. J. Cell Sci. 2003, 116, 1305–1318.
  79. Liang, L.; Fang, J.; Han, X.; Zhai, X.; Song, Y.; Lu, Y.; Zhang, Q.; Ma, R. Prognostic Value of CEA, CA19-9, CA125, CA724, and CA242 in Serum and Ascites in Pseudomyxoma Peritonei. Front. Oncol. 2021, 11, 594763.
  80. Li, B.; Li, Y.; Li, C.; Yang, J.; Liu, D.; Wang, H.; Xu, R.; Zhang, Y.; Wei, Q. An ultrasensitive split-type electrochemical immunosensor based on controlled-release strategy for detection of CA19-9. Biosens. Bioelectron. 2023, 227, 115180.
  81. Poruk, K.E.; Firpo, M.A.; Adler, D.G.; Mulvihill, S.J. Screening for Pancreatic Cancer: Why, How, and Who? Ann. Surg. 2013, 257, 17–26.
  82. Badheeb, M.; Abdelrahim, A.; Esmail, A.; Umoru, G.; Abboud, K.; Al-Najjar, E.; Rasheed, G.; Alkhulaifawi, M.; Abudayyeh, A.; Abdelrahim, M. Pancreatic Tumorigenesis: Precursors, Genetic Risk Factors and Screening. Curr. Oncol. 2022, 29, 8693–8719.
  83. Hatzakis, K.D.; Froudarakis, M.E.; Bouros, D.; Tzanakis, N.; Karkavitsas, N.; Siafakas, N.M. Prognostic Value of Serum Tumor Markers in Patients with Lung Cancer. Respiration 2002, 69, 25–29.
  84. Ferrigno, D.; Buccheri, G.; Giordano, C. Neuron-Specific Enolase Is an Effective Tumour Marker in Non-Small Cell Lung Cancer (NSCLC). Lung Cancer 2003, 41, 311–320.
  85. Gazdar, A.F.; Carney, D.N.; Nau, M.M.; Minna, J.D. Characterization of Variant Subclasses of Cell Lines Derived from Small Cell Lung Cancer Having Distinctive Biochemical, Morphological, and Growth Properties. Cancer Res. 1985, 45, 2924–2930.
  86. Isgrò, M.A.; Bottoni, P.; Scatena, R. Neuron-Specifi c Enolase as a Biomarker: Biochemical and Clinical Aspects. Adv. Exp. Med. Biol. 2015, 867, 125–143.
  87. Tian, Z.; Liang, C.; Zhang, Z.; Wen, H.; Feng, H.; Ma, Q.; Liu, D.; Qiang, G. Prognostic Value of Neuron-Specific Enolase for Small Cell Lung Cancer: A Systematic Review and Meta-Analysis. World J. Surg. Oncol. 2020, 18, 1–8.
  88. Campagnari, F.; Bombardieri, E.; de Braud, F.; Baldini, L.; Maiolo, A.T. Terminal Deoxynucleotidyl Transferase, Tdt, as a Marker for Leukemia and Lymphoma Cells. Int. J. Biol. Markers 1987, 2, 31–42.
  89. Peng, Y.; Shang, H.; Zheng, Z.; Li, H.; Chen, W.; Xu, J. Self-activation of symbiotic multi-DNA machines transducing exponentially amplified fluorescence for ultrasensitively signaling of terminal deoxynucleotidyl transferase activity. Sens. Actuators B. Chem 2023, 380, 133400.
  90. Sur, M.; AlArdati, H.; Ross, C.; Alowami, S. TdT Expression in Merkel Cell Carcinoma: Potential Diagnostic Pitfall with Blastic Hematological Malignancies and Expanded Immunohistochemical Analysis. Mod. Pathol. 2007, 20, 1113–1120.
  91. Mathewson, R.C.; Kjeldsberg, C.R.; Perkins, S.L. Detection of Terminal Deoxynucleotidyl Transferase (TdT) in Nonhematopoietic Small Round Cell Tumors of Children. Pediatr. Pathol. Lab. Med. J. Soc. Pediatr. Pathol. Affil. Int. Paediatr. Pathol. Assoc. 1997, 17, 835–844.
  92. Chapman, M.H.; Sandanayake, N.S.; Andreola, F.; Dhar, D.K.; Webster, G.J.; Dooley, J.S.; Pereira, S.P. Circulating CYFRA 21-1 Is a Specific Diagnostic and Prognostic Biomarker in Biliary Tract Cancer. J. Clin. Exp. Hepatol. 2011, 1, 6–12.
  93. Zhang, L.; Liu, D.; Li, L.; Pu, D.; Zhou, P.; Jing, Y.; Yu, H.; Wang, Y.; Zhu, Y.; He, Y.; et al. The Important Role of Circulating CYFRA21-1 in Metastasis Diagnosis and Prognostic Value Compared with Carcinoembryonic Antigen and Neuron-Specific Enolase in Lung Cancer Patients. BMC Cancer 2017, 17, 96.
  94. Dal Bello, M.G.; Filiberti, R.A.; Alama, A.; Orengo, A.M.; Mussap, M.; Coco, S.; Vanni, I.; Boccardo, S.; Rijavec, E.; Genova, C.; et al. The Role of CEA, CYFRA21-1 and NSE in Monitoring Tumor Response to Nivolumab in Advanced Non-Small Cell Lung Cancer (NSCLC) Patients. J. Transl. Med. 2019, 17, 74.
  95. Rapellino, M.; Niklinski, J.; Pecchio, F.; Furman, M.; Baldi, S.; Chyczewski, L.; Ruffini, E.; Chyczewska, E. CYFRA 21-1 as a Tumour Marker for Bronchogenic Carcinoma. Eur. Respir. J. 1995, 8, 407–410.
  96. Dall’Olio, F.G.; Abbati, F.; Facchinetti, F.; Massucci, M.; Melotti, B.; Squadrilli, A.; Buti, S.; Formica, F.; Tiseo, M.; Ardizzoni, A. CEA and CYFRA 21-1 as Prognostic Biomarker and as a Tool for Treatment Monitoring in Advanced NSCLC Treated with Immune Checkpoint Inhibitors. Ther. Adv. Med. Oncol. 2020, 12, 1758835920952994.
More
Video Production Service