Loading

JSM Clinical Oncology and Research

Clinical Relevance of Tumor Markers in Gastrointestinal Cancers: A Critical Analysis and Literature Review

Review Article | Open Access | Volume 12 | Issue 1

  • 1. Science Center Donaustadt, Austria
  • 2. Medizinsiche Abteilung Klinik Donaustadt, Austria
+ Show More - Show Less
Corresponding Authors
Zottl Jakob, Science Center Donaustadt, Austria
ABSTRACT

Objective: The objective of this article is to offer current and evidence-based recommendations for the application of the most prominent tumor markers in various gastrointestinal cancers, especially Colorectal Carcinoma (CRC), pancreatic cancer and Hepatocellular Carcinoma (HCC). We focused on the most used tumor markers being used for diagnostic and therapeutic purposes for intestinal malignomas being Carcino Embryonic Antigen (CEA), Carbohydrate Antigen 19-9 (CA 19-9) and Alpha-FetoProtein (AFP) . We will describe their constraints and explain the usage in clinical practice and their significance in primary diagnosis, therapy monitoring and the early detection of recurrences, to enhance the quality of multidisciplinary care of cancer patients.

Conclusion: Tumor markers have a clear role in clinical practice. In situations where imaging results are inconclusive, tumor markers offer additional confirmation of recurrence and prove valuable in postoperative surveillance and assessing the response to various therapeutic modalties. Due to their limited positive predictive value, specificity and sensitivity, they are not suitable for screening purposes. Knowledge of this markers is crucial to enhance the multidisciplinary care and treatment coordination for cancer patients. Therefore they should be included into future guidelines, surveillance-algorythms and staging-programmes for certain tumor-stages.

Keywords

• Tumor markers

• Carcinoembryonic antigen

• Carbohydrate antigen 19-9

• Alpha- fetoprotein

• GI Cancers

• Colorectal carcinoma

• Pancreatic cancer

• Hepatocellular carcinoma

• Biosensors

• Biomarkers

CITATION

Jakob Z, Günther SC, Marie-Christine S, Sebesta C (2024) Clinical Relevance of Tumor Markers in Gastrointestinal Cancers: A Critical Analysis and Literature Review. JSM Clin Oncol Res 12(1): 1073.

ABBREVATIONS

AFP: Alpha-Fetoprotein; AJCC: American Joint Comitee on Cancer; ASCO: American Society of Clinical Oncology; Au: Gold; CA 125: Cancer Antigen 125; CA 72-4: Cáncer Antigen 72-4; CA19-9: Carbohydrate Antigen 19-9; CEA: Carcinoembryonic Antigen; CRC: Colorectal Cancer; CT: Computed Tomography; ESMO: European Society of Clinical Oncology; Fe3 O4 : Iron (III) Oxide; GI: Gastrointestinal; HCC: Hepatocellular Carcinoma; IARC: International Agency for Research on Cancer; LDH: Lactate Dehydrogenase; NCCN: National Co0.mprehensive Cancer Network; NSE: Neuron-Specific Enolase; PET: Positron Emisson Tomography; SEER: The Surveillance Epidemiology and End Results; TNM: Tumor Nodes Metastases 

INTRODUCTION

When taking a look at numbers of new cases and deaths, gastrointestinal cancers represent highest numbers in both categories. Colorectal Cancer (CRC) is still the most dominant of them, with approximately 1.9 million new cancer diagnoses in 2020, making it the third most prevalent cancer after lung and breast cancer. In the same year The International Agency for Research on Cancer (IARC) reported 1.1 million new cases of gastric cancer, 900 000 new cases of liver cancer, 600 000 new cases of esophageal cancer and 500 000 new cases of pancreatic cancer globally [1].

In the era of personalized medicine and individualized therapy options in oncology, predictive markers measuring presence or absence of response to specific treatment or interventions are widely used in practice today [2]. Currently, there are several serum-based tumor markers available for diagnosis, prognosis and monitoring of GI cancer, including CarcinoEmbryonic Antigen (CEA), Carbohydrate Antigen 19-9 (CA19-9) and AlphaFetoprotein (AFP) being the most researched, as well as a huge number of other markers like Cancer-Antigen 72-4 (CA 72-4), Cancer Antigen 125 (CA 125), Chromogranin A, Neuron-Specific Enolase (NSE) and Lactate Dehydrogenase (LDH) [3-5]. With CEA, CA 19-9 and AFP being the most researched, most used in clinical practice and best available tumor markers, this article focused its research and literature review on those markers [5,6].

Traditionally, they have been deployed in clinical practice, for monitoring disease response. This involves evaluating treatment effectiveness by observing decrease of a marker, which was previously elevated in primary diagnosis [7]. Several research studies have highlighted the clinical importance of each tumor marker [8-10]. However with limited sensitivity and specificity, especially adding low positive predictive values for applying markers in asymptomatic patients, the use of those markers for screening purposes is still limited and does not correspond to the original objective [11,12].

In this article we will review serum tumor markers used in the most common gastrointestinal tumors, being colorectal carcinoma, pancreatic cancer and hepatocellular carcinoma, according to their normal and pathological values, sensitivity and specificity in their relevance as supplementary information to imaging diagnostics and their ability for serving as screening tools. Additionally, their significance in monitoring treatment success and anticipating recurrences will be discussed and backed up by current literature. Special focus was given to reviews including the few relevant systematic reviews, large studies and clinical guidelines published by expert panels.

TUMOR MARKERS IN GI CANCERS

Definition and function of tumor markers

Traditionally, the term tumor marker is used to describe substances, which are found in the blood as well as other body fluids such as urine, pleural fluid or peritoneal fluid, that are either produced by tumor cells themselves or by other cells in the body as an expression of the presence of a malignancy [13]. Key qualities characterizing an ideal tumor marker are exhibition of high specificity for a particular type of tumor, allowing early detection before clinical diagnosis and demonstrating high sensitivity to minimize false positive results. Furthermore, it should have a short half-life, have reliable correlation with tumor size and be cheap enough for screening in targeted populations [14]. Table 1 gives an overview of common markers and their associated gastrointestinal malignancies [5,15,16].

Table 1: Common tumormarkers and their associated malagnacies.

Marker

Associated Cancers

Normal Value

 

CEA

Colorectal cancer Pancreatic cancer

< 2.5 ng/ml in non- smokers

<5 ng/ml in smokers

 

CA 19-9

Pancreatic adenocarcinoma Cholangiocarcinoma Colorectal cancer

 

0-37 U/ml

CA 125

Pancreatic ductal adenoma

0-35 U/ml

AFP

Hepatocellular carcinoma

0-10 ng/ml

CA 72-4

Gastric cancer

< 6 U/ml

Carcino Embryonic Antigen (CEA)

CEA is released from the apical surface of mature columnar cells into the gut lumen and has restricted expression in normal adult tissue. Due to loss of polarity and absence of basal lamina CEA is spread out around the cell surface and accumulates at high levels in the blood [17].

Being a high glycosylated cell surface glycoprotein, it consist of amino-terminal domain with a processed leader sequence, three internal domains, as well as a short 27 amino acid hydrophobic carboxy terminal-domain. CEA’s amino acid sequence shows notable similarity to proteins in the immunoglobulin superfamily, which includes molecules involved in intercellular recognition process, such as immunoglobulins, T cell receptors, growth factor receptors and intercellular adhesion molecules like N-Cam [18]. Studies propose, that CEA and its family member CEACAM6 are upregulated in cells retaining division potential and can result in a gradual decline of cellular polarization, distortion of tissue architecture as well as inhibition of differentiation [19].

To determine situations in which tumor markers have proven clinical validity, clinical practice guidelines should be taken for scientific evidence [13]. Comparing guidelines, elevated preoperative CEA levels do not fall within the clinicopathological factors that categorize stage II colon cancer as ‘high risk’, according to American Society of Clinical Oncology (ASCO) [20] and the National Comprehensive Cancer Network (NCCN) [21]. Despite not being updated since 2006, ASCO published recommendations for the use of tumor markers in gastrointestinal cancers. Guidelines recommend ordering CEA preoperatively for patients if it can aid in determining extend of the cancer and assist in planning surgical treatments [22]. Guidelines published by the European Group on Tumor Markers in 2014 follow a similar approach recommending measuring preoperative CEA levels in newly diagnosed CRC patients. Guidelines published by the European Society of Clinical Oncology (ESMO) follow a different approach in considering high preoperative CEA levels as a minor prognostic parameter for stage II disease. Nevertheless all Guidelines suggest CEA levels should be evaluated before surgery and examined regularly during the post-operative period to help early detection of metastatic disease. However, due to low specificity and sensitivity the level of serum-CEA are not sufficient for colon cancer diagnosis in absence of a confirmatory tumor biopsy [23].

Comparing concrete preoperative serum levels, several studies show numbers >5.0 ng/mL to negatively affect survival outcomes regardless of the tumor’s stage [24-28]. For example a study published by Wiratkapun et al. showed significantly higher percentages of patients developing metastasis having CEA levels above 5 ng/ml in comparison to patients with lower numbers. This study also indicated numbers higher than 15 ng/ml to be a significant prognostic indicator for disease-free survival [29]. Several studies showed similar effects for patients with stage II disease [30-32]. Another study by Thirunavukarasu analyzed 17 910 colon cancer patients at any stage, which were reported to SEER database in 2004. Those patients with elevated preoperative CEA level had significantly higher risk of overall mortality (HR for death 1.60, 95% CI 1.46-1.76) [27]. The largest study to date examining correlation between elevated CEA levels and overall survival in CRC patients in 2016 included 137 381 patients and was published by Adan Z. et al. This study showed an increase hazard of death by 62% for patients showing elevated preoperative CEA levels in comparison to patients with normal CEA levels [33].

With data indicating high levels of preoperative CEA having prognostic significance, some experts suggest integrating preoperative CEA levels into the traditional TNM staging system for colon cancer [34]. The 2017 revision of the system does not directly use serum CEA levels for assigning stages but suggest gathering data for its prognostic value and for postoperative monitoring to detect recurrences [35,36].

With 50% of patients presenting with nonmetastatic disease and manifesting metastatic disease, as well as approximately 35% of patients presenting with metastatic disease at diagnosis, studies show benefit in early detection of metastases as metastasectomy improves patient’s survival [5,37] With liver being the most prominent organ for metastasis, overexpression of CEA can be used as a tumor marker after cancer therapy or surgery in cancer patients [38]. As recommended by NCNN, postoperative CEA levels should be measured every 3 to 6 months for at least 2 years, then every 6 months for a total of 5 years for tumors higher than stage II. Patients showing serial CEA elevation should get workup-treatment consisting of physical examination, colonoscopy and even chest/abdominal/pelvic CT with contrast [39].

Guidelines published by ASCO prior to 2018 follow similar approaches for stage II and III disease [22,40]. ESMO guidelines recommend determination of CEA levels every 3-6 months for 3 years, as well as every 6-12 months at years 4 and 5 after surgery combined with CT scan of chest and abdomen with colonoscopy A systematic review published in 2017 by Shinkins et al. recommended testing frequency should be increased to monthly for 3 months and then every 2 months with threshold for investigating being 10 µg/l after the second CEA test [41]. However, a Cochrane Database systematic review published in 2015 concluded CEA being insufficiently sensitive used alone, therefore it is crucial to impliment CEA monitoring together with another diagnostic method to avoid missed cases [42]. Despite checking CEA levels, periodical clinical visits as well as CT and endoscopy should be used for surveillance for CRC [16,43]. On the other hand, there are studies showing little or no minimal benefit using CEA for surveillance [44,45], as well as claiming, that only 30% of all CRC release CEA in early states [46].

Another clinical advantage measuring CEA levels consists in the prediction of response to chemotherapy and in monitoring progression under treatment of advanced CRC patients undergoing chemotherapy [5]. International guidelines also recommend following this strategy in advanced tumor stages, recommending measurement of CEA at the start of treatment for metastatic disease and every 1 to 3 months during active treatment [22,40]. A study published by de Haas et al. compared correlation between tumor marker response and radiological response in form of computed tomography. Patients with isolated liver metastasis showed accordance in >90% of cases, patients with stable diseases in 94% of cases and patients showing radiological evidence of disease progression showed correlation in 95% of cases [40,47].

It is necessary to mention, that rises in CEA levels may occur in some assays, being falsely elevated in the context of cigarette smoking or adjuvant 5-Fluouracil (FU) treatment during the first 4 to 6 weeks [22,46].

Carbohydrate antigen 19-9

As a mucinous marker, glycoprotein CA 19-9, also termed Sialyl Lewis [48], is produced by various normal cells in the pancreas, bile ducts, stomach and saliva glands and presented in small amounts in the bloodstream. Because of alteration in processes that regulate the passage of CA 19-9 into the bloodstream, levels tend to increase during cancer development as well as certain inflammatory conditions like pancreatitis and a number of other pancreato-biliary conditions [49].

Being a tetrasaccharid, carbohydrate Antigen 19-9 is synthazised by glucosyltransferases which sequentially bind the monosaccharide precursors onto both N-linked and O-linked glykans. The Lewis blood group system consist of a set of fucosylated glycosphingolipids synthesized by exocrine epithelial cells. These are subsequently adsorbed onto the surface of erythrocytes defining their Lewis phonotype and circulating as red blood cells in body fluids [49].

It has been widely recognized, that glycan structures on the cell surface undergo significant alterations during malignant transformation. These changes are attributed to a process referred to as ‘incomplete synthesis’ of complex carbohydrate determinants, leading to the expression of structurally less complex carbohydrate molecules [49].

Clinical guidelines published by ESMO in 2015 state that CA 19-9 is not effective as a primary diagnostic marker for pancreatic cancer, despite an elevation of the marker can be seen in 80% of patients with advanced disease [50]. Data indicate measuring preoperative CA 19-9 levels correlate with staging by American Joint Comitee on Cancer (AJCC) and resectability. Therfore, recent guidelines published by NCCN recommend the use of CA 19-9 as a diagnostic marker, adding insights to the staging process [51].

In order to represent an accurate baseline, CA 19-9 levels should be best performed after biliary decompression is completed by anymeans and bilirubin is normal or back to baseline. Hence, Guidelines suggest measuring CA 19-9 levels after neoadjuvant therapy, pre-surgery, post-surgery before starting adjuvant treatment and during surveillance [51].

Using serum CA 19-9 as a screening tool was observed in two large studies [52,53], concluding, that screening in asymptomatic individuals using CA 19-9 as a marker for malignant disease showed no efficiency, due to its low positive predictive value [54]. In a large study involving more than 70 000 asymptomatic individuals, serum CA 19-9 level >37 U/L had a Positive Predicative Value (PPV) of only 0.9 percent [55]. Even when using elevated CA 19-9 for screening symptomatic patients, sensitivity and specificity are approximately 79-81% and 82-80% [56]. A metanalysis published by Zhang et al in 2015 comparing diagnostic power of CA 19-9 to other markers such as CA 242 and CEA showed CA 19-9 to have the highest sensitivity and CA 242 to have highest specificity, indicating that combination of both markers in one test pattern could increase sensitivity without impairing specificity in diagnosis of pancreatic cancer [57].

As a prognostic factor, CA 19-9 has notable importance as well as an indicator to assess the progression of disease and potentially influence treatment choices. Several studies show that the extent of CA 19-9 elevation both at the initial diagnosis and after surgery is linked to long-term survival [58-60]. ESMOguidelines recommend taking preoperative serum levels of CA 19-9 >500 IU/ml as indication of poor prognosis after surgery [50]. Furthermore studies suggest that normalization or decrease in postoperative CA 19-9 levels by >20% to 50% from baseline after surgery or chemotherapy correlate with prolonged survival [61]. A study published by Ferrone et al. stated decrease in CA 19-9 as well as CA 19-9 value less than 200 IU/ml as significant predictors in survival for patients with pancreatic adenocarcinoma even after adjusting for stage [62]. Taking a look at the chronological relationship between CA 19-9 elevation and radiographic recurrence, data show that CA 19-9 disappointingly provides only poor positive predictive value (average 35%) but high negative predictive value (average 92%) for recurrence at 6-months intervals. However, data also shows CA 19-9 being able to predict prognosis at each survival interval and precede both clinical and radiological signs [16,63].

A study reviewing 491 patients undergoing staging laparoscopy for radiographically resectable pancreatic adenocarcinoma assessed association between preoperative CA 19-9 levels with presence of subradiographic unresectable disease. Results showed that values greater than 130 U/ml was a predictor of tumor unresectability and indicated preoperative CA 19-9 levels to allow surgeons to better select patients for staging laparoscopy [64].

In accordings with these findings, monitoring CA 19-9 levels can also be used to predict prognosis and incidence of recurrence after neoadjuvant therapy [16]. A multicenter case-control study by Aoki et al. revealed CA 19-9 level ≤ 103 IU/ml to be a significant predictor of overall survival with better prognosis and lower hepatic recurrence after surgery for patients receiving neoadjuvant therapy [65]. For adjuvant chemotherapy measuring serum CA 19-9 levels could also have potential clinical utility in predicting outcome and response [66].

It is important to mention, that 10% of the population lack the enzyme needed for epitope production of Sialyl Lewis antigen called 1,4-fucosyl transferase. Due to the fact, that these patients are unable to produce CA 19-9, measurement of this marker will provide falsely negative results [56].

Alpha-fetoprotein

Deriving from embryonic endoderm tissue cells, AFP is released into fetal bloodstream, with mature hepatocytes being unable to release this glycoprotein. Through malignant transformation liver cancer cells are able to produce AFP again making it the most commonly used tumor marker for Hepatocellular Carcinoma (HCC) [67,68].

The AFP gene is part of the albumin gene family forming a multigene cluster distinguished by their affinity for the lectin Lens culinaris agglutinin: AFP-L1, AFP-L2, and AFP-L3. The regulation of AFP expression primarily occurs at the transcriptional level with the gene having an upstream region, including a tissuespecific promoter, three idependent enhancers and two silencer regions. According to preclinical studies, it seems, that in adult cells, the expression of AFP is inhibited at the promoter and two enhancers through the involvement of corepressors and methylated histones [69].

AFP is mainly snythosized during embryonic development by fetal liver, visceral endoderm of the yolk sac and reaches its maximal value in human fetal blood at 12-16 week of gestitation. In typical circumstances, the concentration of AFP in the blood of adult humans is approximately 5 to 10 ng/ml [70].

Using a threshold value of 20ng/ml in patients with cirrhosis sensitivity and specificity for detecting HCC vary from 41-65% and 80-94% respectively [71]. Due to its low positive predictive value and the fact that AFP levels are normal in about 30 to 40% of patients with HCC, AFP alone is not recommended as a screening tool, nor as surveillance for patients with risk for developing HCC [72]. Due to its weakness as a screening tool, clinical practice guidelines recommend using ultrasound with or without AFP measurement for patients at risk [73-75].

Several studies highlight the importance of using alphafetoprotein as a diagnostic tool for monitoring prognosis and survival in patients with a proven HCC diagnosis [76,77]. Data shows significantly lower survival rate for patients showing AFP levels of ≥400 IU/ml in comparison to patients with lower levels [78], with one study showing patients with tumor sizes >5cm and AFP levels >1000 to have an 82% incidence of vascular invasion [79]. Furthermore studies showed that HCC patients with higher AFP levels are likely to have greater tumor size, bilobar involvement and portal vein thrombosis [80].

Another important indication is that AFP can be used for surveillance in patients after receiving liver transplantation [72]. Data indicate AFP progression of more than 15µg/l per month to be a major predictive factor for tumor recurrence, as well as for poor survival after transplantation [81]. A comparative study by Merani et al. showed that among patients who had initial AFP levels >400ng/ml at time of listing, those who achieved reduction in AFP to 400 or lower experienced significantly better overall survival than patients who did not succeed to this reduction [82]. Data also prove AFP levels to predict recurrence after transplantation, with AFP levels >200ng/ml being associated with higher relapse rates in patients after liver transplantation, showing other risk factors such as presence of vascular invasion or satellite nodules. Therefore, including measurement of AFP levels has been suggested to be included when selecting patients for inclusion on liver transplant lists, as well as a surrogate marker of poor pathological conditions after transplantation [72,83].

Predicting radiological response and survival of patients undergoing systemic chemotherapy, data indicate that AFP response after receiving systemic therapy correlates with better survival rates and tend to identify patients with stable disease more likely. Furthermore AFP-response is clearly associated with radiographic response and qualifies as a prognostic factor for progression-free survival and overall survival [84,85]. Besides chemotherapy, measurement of AFP levels also provides prognostic value for surveillance of antiangiogenic drugs like bevacizumab [86], sorafenib [87] or ramucirumab [88], as well as tyrosine kinase-inhibitors such as cabozantinib [89].

Improving diagnostic accuracy

Despite several markers showing benefits when using in multimodal diagnostic approaches, combination of multiple biomarkers is necessary to improve the accuracy of diagnostic tests [90]. Due to the fact, that relying on a single biomarker for diagnosis may lack sufficient accuracy, it is increasingly prevalent in medical research and practice to conduct multiple biomarker tests on individuals and combine corresponding measurements into a unified score [91,92]. Using more diagnostic markers simultaneously increases the chance of one marker showing higher sisnsitivity [93]. In recent years the addition of nanomaterials for detection of biomarkers correlating with cellular alterations has attracted tremendous attention. There have been ongoing developments of nanomaterials including carbon nanotubes, graphene, Au, and Fe3 O4 -based biosensors being used in early disease detection and diagnostics of lung, prostate, breast and colon cancer [94]. Despite having a higher diagnostic accuracy, imaging methods such as MRI, CT and PETCT are usally very expensive and are often times inefficient for early stage cancer detection as these methods depend on phenotypic charcteristics of the tumor [95,96]. Including biosensors alongside tumor markers into the diagnostic process could be a more cost-effective and non-invasive methode, with a fast response due to the ability of direct assement of the physiological fluids [97]. Furthermore protein and microRNA based biomarkers and corresponding biosensors are becoming a promising tool forearly diagnosis of GI cancers and propose another diagnostic paramaeter to add to improve diagnostic accuracy [97,98].

CONCLUSIONS

Tumor markers play an important role in today’s multimodal therapeutic process in modern oncology, which is mainly driven by precision medicine and innovative treatment strategies. This review aimed to give an overview of three most studied and used tumor markers in clinical practice being CEA, CA 19-9 and AFP. Although some issues related to their importance have not been fully evaluated, the significance of the markers described in this review is of clear use in everyday clinical practice in specific situations.

Clinical guidelines recommend preoperative CEA measurement to better understand disease progression and support treatment planning. Studies show, that elevated preoperative CEA levels, especially above 5.0 ng/ml are associated with poor prognosis and reduced survival rates. Therefore, measurement of CEA levels should be a part of future recommendations by clinical practice guidelines, as well as including high levels into high-risk features for stage II colon cancer. It is important to mention, that high CEA levels alone do not provide definitive diagnosis due to its low sensitivity and specificity, and complementary diagnostic methods are necessary. However, testing CEA levels regularly in the years following surgery can be useful in postoperative monitoring. CEA plays also a role in assessing response rates to chemotherapy and monitoring treatment process.

While using CA 19-9 as a diagnostic marker for primary diagnostics is not recommended due to its limited specificity and sensitivity, it holds significant prognostic significance and can serve as an indicator to assess disease severity and potentially influence treatment. Attempts of using CA 19-9 as a tool for screening in asymptomatic induvials have yielded into negative results, due to its low positive predictive value. Even in symptomatic individuals its sensitivity and specificity are suboptimal. As a prognostic factor elevated levels of CA 19-9 at diagnosis and post-treatment are associated with poor longterm survival, while a decrease in CA 19-9 levels after surgery correlates with improved outcome. Studies showed that CA 19-9 can also predict recurrence and prognosis at various intervals, even preceding clinical and radiological signs. Furthermore, CA 19-9 can aid in patient selection for staging laparoscopy and has potential utility predicting outcomes and responses to neoadjuvant and adjuvant therapy.

While AFP alone is not recommended for a screening tool, due to its low positive predictive value and the fact that a significant portion of HCC patients remain undetected, it holds immense value in various clinical aspects of HCC management. Elevated AFP levels are associated with larger tumor size, bilobar involvement, specific tumor types, as well as vascular invasion, all of which are critical factors influencing patient’s outcome. Furthermore, AFP proves value in post-liver transplantation setting, with progressive AFP elevation correlating with poor survival and as predictive factor for tumor recurrence. Data indicate that patients achieving lower AFP levels after transplantation tend to experience better overall survival. In HCC-Patients AFP offers insight into response to chemotherapy and antiangiogenic drugs, correlating with radiographic response, ProgressionFree Survival (PFS) and even Overall Survival (OS) in patients undergoing systemic therapy.

A remarkable study done in 2021 analyzing diagnostic and prognostic value of CEA and CA 19-9 in CRC showed 69% of patients with CEA and 66% with CA 19-9 levels greater than 200 had significantly shorter 5-year overall survival, as well as patients with both tumormarkers increased, showed remarkably shorter 5-year survival rate of 23% [99]. Furthermore, patients with both markers elevated had shortest recurrence-free survival rates of 44% and also shorter rates when only one marker was elevated (65%) in comparison to patients showing no elevation (79%) [99]. This emphasizes the importance of including measurement of these tumor markers from the onset of diagnosis, as well as throughout the treatment process.

Overviewing current and past data published on tumor markers, it can be concluded, that due to their low positive predictive value, as well as specificity and sensitivity, they should not be used for screening purposes. However, data indicate their role in clinical practice clearly. Tumor markers may provide additive confirmation for recurrence in doubtful situations, where imaging is not absolutely conclusive. Furthermore they are a valuable add- on- information in the situation of postoperative surveillance and in the assesemant of the response to neo-adjuvant, adjuvant and palliative treatment of all available therapy modalities, such as classical chemotherapy, targeted treatments, immune- therapy, radiation therapy or a combination of all the mentioned options.

Therefore, future clinical guidelines should consequently and strictly evidence- based include tumor markers into the surveillance- algorithms, consider them as prognostic parameters to estimate survival, include them into the TNM staging in defined stages and recommend the optimal times for the repeated determination, in order to characterize the further course of the disease as well as to estimate the individual prognosis of the patient more precisely.

Conflicts of Interest: There was no conflict of interest.

REFERENCES
  1. International Agency for Research on Cancer. Estimated Number of New Cases and Deaths of Cancer in 2020. 2020.
  2. Lahoud RM, O’Shea A, El-Mouhayyar C, Atre ID, Eurboonyanun K, Harisinghani M. Tumour markers and their utility in imaging of abdominal and pelvic malignancies. Clin Radiol. 2021; 76: 99–107.
  3. David P, Mittelstädt A, Kouhestani D, Anthuber A, Kahlert C, Sohn K, et al. Current applications of liquid biopsy in gastrointestinal cancer disease-from early cancer detection to individualized cancer treatment. Cancers (Basel). 2023; 15: 1924.
  4. Isgrò MA, Bottoni P, Scatena R. Neuron-specific enolase as abiomarker: Biochemical and Clinical Aspects. 2015; 125–143.
  5. Faria SC, Sagebiel T, Patnana M, Cox V, Viswanathan C, Lall C, et al. Tumor markers: Myths and facts unfolded. Abdominal Radiology. 2019; 44: 1575–1600.
  6. Holdenrieder S, Pagliaro L, Morgenstern D, Dayyani F. Clinically meaningful use of blood tumor markers in oncology. Biomed Res Int. 2016; 1–10.
  7. Yang XQ, Chen C, Wang FB, Peng CW, Li Y. Preoperative serum carcinoembryonic antigen, carbohydrate antigen19-9 and carbohydrate antigen 125 as prognostic factors for recurrence-free survival in colorectal cancer. Asian Pac J Cancer Prev. 2011; 12: 1251–1256.
  8. Lee JH, Lee SW. The roles of carcinoembryonic antigen in liver metastasis and therapeutic approaches. Gastroenterol Res Pract. 2017; 1–11.
  9. Ballehaninna UK, Chamberlain RS. The clinical utility of serum CA 19-9 in the diagnosis, prognosis and management of pancreatic adenocarcinoma: An evidence based appraisal. J Gastrointest Oncol. 2012; 3: 105–119.
  10. Bagaria B, Sood S, Sharma R, Lalwani S. Comparative study of CEA and CA19-9 in esophageal, gastric and colon cancers individually and in combination (ROC curve analysis). Cancer Biol Med. 2013; 10: 148–157.
  11. Duffy MJ. Use of biomarkers in screening for cancer. EJIFCC. 2010; 21: 1–12.
  12. Duffy MJ. Clinical uses of tumor markers: A critical review. Crit Rev Clin Lab Sci. 2001; 38: 225–262.
  13. Filella X, Rodríguez-Garcia M, Fernández-Galán E. Clinical usefulness of circulating tumor markers. Clinical Chemistry and Laboratory Medicine (CCLM). 2023; 61: 895–905.
  14. Sharma S. Tumor markers in clinical practice: General principles and guidelines. Indian Journal of Medical and Paediatric Oncology. 2009; 30: 1–8.
  15. Passek K, Ohlendorf D, Wanke E, Bendels MH, Oremek GM, Ziolkowski P, et al. Der Tumormarker CA 72-4. Zentralbl Arbeitsmed Arbeitsschutz Ergon. 2018; 68: 38–40.
  16. Lahoud RM, O’Shea A, El-Mouhayyar C, Atre ID, Eurboonyanun K, Harisinghani M. Tumour markers and their utility in imaging of abdominal and pelvic malignancies. Clin Radiol. 2021; 76: 99–107.
  17. Hammarström S. The Carcinoembryonic Antigen (CEA) family: Structures, suggested functions and expression in normal and malignant tissues. Semin Cancer Biol. 1999; 9: 67–81.
  18. Benchimol S, Fuks A, Jothy S, Beauchemin N, Shirota K, Stanners CP. Carcinoembryonic antigen, a human tumor marker, functions as an intercellular adhesion molecule. Cell. 1989; 57: 327–334.
  19. Ilantzis C, Demarte L, Screaton RA, Stanners CP. Deregulated expression of the human tumor marker CEA and CEA family member CEACAM6 disrupts tissue architecture and blocks colonocyte differentiation. Neoplasia. 2002; 4: 151–63.
  20. Baxter NN, Kennedy EB, Bergsland E, Berlin J, George TJ, Gill S, et al. Adjuvant therapy for stage II colon cancer: ASCO guideline update. Journal of Clinical Oncology. 2022; 40: 892–910.
  21. National Comprehensive Cancer Network (NCCN). NCCN clinical practice guidelines in oncology.
  22. Locker GY, Hamilton S, Harris J, Jessup JM, Kemeny N, Macdonald JS, et al. ASCO 2006 update of recommendations for the use of tumor markers in gastrointestinal cancer. Journal of Clinical Oncology. 2006; 24: 5313–27.
  23. Argilés G, Tabernero J, Labianca R, Hochhauser D, Salazar R, Iveson T, et al. Localised colon cancer: ESMO clinical practice guidelines for diagnosis, treatment and follow-up. Annals of Oncology. 2020; 31: 1291–305.
  24. Wolmark N, Fisher B, Wieand HS, Henry RS, Lerner H, Legault-Poisson S, et al. The prognostic significance of preoperative carcinoembryonic antigen levels in colorectal cancer. Ann Surg. 1984; 199: 375–382.
  25. Meling GI, Rognum TO, Clausen OPF, Børmer O, Lunde OC, Schlichting E, et al. Serum carcinoembryonic antigen in relation to survival, dna ploidy pattern, and recurrent disease in 406 colorectal carcinoma patients. Scand J Gastroenterol. 1992; 27: 1061–1068.
  26. Park IJ, Choi GS, Lim KH, Kang BM, Jun SH. Serum carcinoembryonic antigen monitoring after curative resection for colorectal cancer: clinical significance of the preoperative level. Ann Surg Oncol. 2009; 16: 3087–3093.
  27. Thirunavukarasu P, Sukumar S, Sathaiah M, Mahan M, Pragatheeshwar KD, Pingpank JF, et al. C-stage in colon cancer: Implications of carcinoembryonic antigen biomarker in staging, prognosis, and management. jnci: journal of the national cancer institute. 2011; 103: 689–697.
  28. thirunavukarasu p, talati c, munjal s, attwood k, edge sb, francescuttiv. Effect of incorporation of pretreatment serum carcinoembryonic antigen levels into ajcc staging for colon cancer on 5-year survival. JAMA Surg. 2015; 150: 747.
  29. Wiratkapun S, Kraemer M, Seow-Choen F, Ho YH, Eu KW. High preoperative serum carcinoembryonic antigen predicts metastatic recurrence in potentially curative colonic cancer. Dis Colon Rectum. 2001; 44: 231–235.
  30. Sun LC, Chu KS, Cheng SC, Lu CY, Kuo CH, Hsieh JS, et al. Preoperative serum carcinoembryonic antigen, albumin and age are supplementary to UICC staging systems in predicting survival for colorectal cancer patients undergoing surgical treatment. BMC Cancer. 2009; 9: 288.
  31. Huh JW, Oh BR, Kim HR, Kim YJ. Preoperative carcinoembryonic antigen level as an independent prognostic factor in potentially curative colon cancer. J Surg Oncol. 2010; 101: 396–400.
  32. Thirunavukarasu P, Sukumar S, Sathaiah M, Mahan M, Pragatheeshwar KD, Pingpank JF, et al. C-stage in colon cancer: Implications of carcinoembryonic antigen biomarker in staging, prognosis, and management. JNCI: Journal of the National Cancer Institute. 2011; 103: 689–697.
  33. Becerra AZ, Probst CP, Tejani MA, Aquina CT, González MG, Hensley BJ, et al. Evaluating the prognostic role of elevated preoperative carcinoembryonic antigen levels in colon cancer patients: results from the national cancer database. Ann Surg Oncol. 2016; 23: 1554– 1561.
  34. . compton cc, fielding lp, burgart lj, conley b, cooper hs, hamilton sr, et al. Prognostic factors in colorectal cancer. Arch Pathol Lab Med. 2000; 124: 979–994.
  35. Jessup JM, Goldberg RM, Asare EA, et al. Colon and Rectum. In: Amin MB, editor. AJCC Cancer Staging Manual, 8th ed. Chicago: AJCC; 2017. 251.
  36. AJCC Cancer Staging Manual, 7th ed. Edge SB, Byrd DR, Compton CC, et al. editors. New York : Springer; 2010. 143.
  37. Piawah S, Venook AP. Targeted therapy for colorectal cancer metastases: A review of current methods of molecularly targeted therapy and the use of tumor biomarkers in the treatment of metastatic colorectal cancer. 2019; 125: 4139–4147.
  38. Lee JH, Lee SW. The roles of carcinoembryonic antigen in liver metastasis and therapeutic approaches. Gastroenterol Res Pract. 2017; 1–11.
  39. Benson AB, Venook AP, Al-Hawary MM, Cederquist L, Chen YJ, Ciombor KK, et al. NCCN Guidelines Insights: Colon cancer, Version 2.2018. Journal of the National Comprehensive Cancer Network. 2018; 16: 359–369.
  40. Duffy MJ, Lamerz R, Haglund C, Nicolini A, Kalousová M, Holubec L, et al. Tumor markers in colorectal cancer, gastric cancer and gastrointestinal stromal cancers: European group on tumor markers 2014 guidelines update. Int J Cancer. 2014; 134: 2513–2522.
  41. Shinkins B, Nicholson BD, James T, Pathiraja I, Pugh S, Perera R, et al. What carcinoembryonic antigen level should trigger further investigation during colorectal cancer follow-up? A systematic review and secondary analysis of a randomised controlled trial. Health Technol Assess (Rockv). 2017; 21: 1–60.
  42. Nicholson BD, Shinkins B, Pathiraja I, Roberts NW, James TJ, Mallett S, et al. Blood CEA levels for detecting recurrent colorectal cancer. Cochrane Database of Systematic Reviews. 2015.
  43. Liu SL, Cheung WY. Role of surveillance imaging and endoscopy in colorectal cancer follow-up: Quality over quantity? World J Gastroenterol. 2019; 25: 59-68.
  44. Duffy MJ. Carcinoembryonic antigen as a marker for colorectal cancer: is it clinically useful? Clin Chem. 2001; 47: 624-30.
  45. Ohlsson B, Breland U, Ekberg H, Graffner H, Tranberg KG. Follow-up after curative surgery for colorectal carcinoma. Dis Colon Rectum. 1995; 38: 619-26.
  46. Moertel CG, Fleming TR, Macdonald JS, Haller DG, Laurie JA, Tangen C. An evaluation of the carcinoembryonic antigen (CEA) test for monitoring patients with resected colon cancer. JAMA. 1993; 270: 943-7.
  47. de Haas RJ, Wicherts DA, Flores E, Ducreux M, Lévi F, Paule B, et al. Tumor marker evolution: comparison with imaging for assessment of response to chemotherapy in patients with colorectal liver metastases. Ann Surg Oncol. 2010; 17: 1010-23.
  48. Indellicato R, Zulueta A, Caretti A, Trinchera M. Complementary use of carbohydrate antigens lewis a, lewis b, and sialyl-lewis a (ca19.9 epitope) in gastrointestinal cancers: Biological rationale towards a personalized clinical application. Cancers (Basel). 2020; 12: 1509.
  49. Scarà S, Bottoni P, Scatena R. CA 19-9: Biochemical and clinical aspects. 2015; 247-260.
  50. Ducreux M, Cuhna ASa, Caramella C, Hollebecque A, Burtin P, Goéré D, et al. Cancer of the pancreas: ESMO Clinical practice guidelines for diagnosis, treatment and follow-up. Annals of Oncology. 2015; 26: 56–68.
  51. National Comprehensive Cancer Network (NCCN). NCCN clinical practice guidelines in oncology.
  52. Homma T, Tsuchiya R. The study of the mass screening of persons without symptoms and of the screening of outpatients with gastrointestinal complaints or icterus for pancreatic cancer in japan, using ca19-9 and elastase-1 or ultrasonography. Int J Pancreatology. 1991; 9: 119–124.
  53. Kim JE, Lee KT, Lee JK, Paik SW, Rhee JC, Choi KW. Clinical usefulness of carbohydrate antigen 19-9 as a screening test for pancreatic cancer in an asymptomatic population. J Gastroenterol Hepatol. 2004; 19: 182–186.
  54. Poruk KE, Gay DZ, Brown K, Mulvihill JD, Boucher KM, Scaife CL, et al. The clinical utility of CA 19-9 in pancreatic adenocarcinoma: Diagnostic and prognostic updates. Curr Mol Med. 2013; 13: 340–351.
  55. Kim JE, Lee KT, Lee JK, Paik SW, Rhee JC, Choi KW. Clinical usefulness of carbohydrate antigen 19-9 as a screening test for pancreatic cancer in an asymptomatic population. J Gastroenterol Hepatol. 2004; 19: 182–186.
  56. Tsen A, Barbara M, Rosenkranz L. Dilemma of elevated CA 19-9 in biliary pathology. Pancreatology. 2018 ; 18: 862–867.
  57. Zhang Y, Yang J, Li H, Wu Y, Zhang H, Chen W. Tumor markers CA19-9, CA242 and CEA in the diagnosis of pancreatic cancer: a meta-analysis. Int J Clin Exp Med. 2015; 8: 11683–11691.
  58. Kondo N, Murakami Y, Uemura K, Hayashidani Y, Sudo T, Hashimoto Y, et al. Prognostic impact of perioperative serum ca 19-9 levels in patients with resectable pancreatic cancer. Ann Surg Oncol. 2010; 17: 2321–2329.
  59. Abdel-Misih SRZ, Hatzaras I, Schmidt C, Saab TB, Klemanski D, Muscarella P, et al. Failure of normalization of ca19-9 following resection for pancreatic cancer is tantamount to metastatic disease. Ann Surg Oncol. 2011; 18: 1116–1121.
  60. Humphris JL, Chang DK, Johns AL, Scarlett CJ, Pajic M, Jones MD, et al. The prognostic and predictive value of serum CA19.9 in pancreatic cancer. Annals of Oncology. 2012; 23: 1713–1722.
  61. Ballehaninna UK, Chamberlain RS. The clinical utility of serum CA 19-9 in the diagnosis, prognosis and management of pancreatic adenocarcinoma: An evidence based appraisal. J Gastrointest Oncol. 2012; 3 :105–119.
  62. Ferrone CR, Finkelstein DM, Thayer SP, Muzikansky A, Castillo CF del, Warshaw AL. Perioperative ca19-9 levels can predict stage and survival in patients with resectable pancreatic adenocarcinoma. J Clin Oncol. 2006; 24: 2897–2902.
  63. Rieser CJ, Zenati M, Hamad A, Al Abbas AI, Bahary N, Zureikat AH, et al. CA19-9 on Postoperative surveillance in pancreatic ductal adenocarcinoma: predicting recurrence and changing prognosis over time. Ann Surg Oncol. 2018; 25: 3483–3491.
  64. Maithel SK, Maloney S, Winston C, Gönen M, D’Angelica MI, DeMatteo RP, et al. Preoperative ca 19-9 and the yield of staging laparoscopy in patients with radiographically resectable pancreatic adenocarcinoma. Ann Surg Oncol. 2008; 15: 3512–3520.
  65. Aoki S, Motoi F, Murakami Y, Sho M, Satoi S, Honda G, et al. Decreased serum carbohydrate antigen 19–9 levels after neoadjuvant therapy predict a better prognosis for patients with pancreatic adenocarcinoma: A multicenter case-control study of 240 patients. BMC Cancer. 2019; 19: 252.
  66. Humphris JL, Chang DK, Johns AL, Scarlett CJ, Pajic M, Jones MD, et al. The prognostic and predictive value of serum CA19.9 in pancreatic cancer. Annals of Oncology. 2012; 23: 1713–1722.
  67. Wang X, Wang Q. Alpha-fetoprotein and hepatocellular carcinoma immunity. Can J Gastroenterol Hepatol. 2018; 2018: 1–8.
  68. Zhao YJ, Ju Q, Li GC. Tumor markers for hepatocellular carcinoma. Mol Clin Oncol. 2013; 1: 593–598.
  69. Galle PR, Foerster F, Kudo M, Chan SL, Llovet JM, Qin S, et al. Biology and significance of alpha-fetoprotein in hepatocellular carcinoma. Liver Int. 2019; 39: 2214–2229.
  70. Terentiev AA, Moldogazieva NT. Structural and functional mapping of α-fetoprotein. Biochemistry (Moscow). 2006; 71: 120–132.
  71. Hanif H, Ali MJ, Susheela AT, Khan IW, Luna-Cuadros MA, Khan MM, et al. Update on the applications and limitations of alpha-fetoprotein for hepatocellular carcinoma. World J Gastroenterol. 2022; 28: 216–229.
  72. Galle PR, Foerster F, Kudo M, Chan SL, Llovet JM, Qin S, et al. Biology and significance of alpha-fetoprotein in hepatocellular carcinoma. Liver International. 2019; 39: 2214–2229.
  73. Heimbach JK, Kulik LM, Finn RS, Sirlin CB, Abecassis MM, Roberts LR, et al. AASLD guidelines for the treatment of hepatocellular carcinoma. Hepatology. 2018; 67: 358–380.
  74. Omata M, Cheng AL, Kokudo N, Kudo M, Lee JM, Jia J, et al. Asia–Pacific clinical practice guidelines on the management of hepatocellular carcinoma: a 2017 update. Hepatol Int. 2017; 11:  317–370.
  75. Galle PR, Forner A, Llovet JM, Mazzaferro V, Piscaglia F, Raoul JL, et al. EASL Clinical Practice Guidelines: Management of hepatocellular carcinoma. J Hepatol. 2018; 69: 182–236.
  76. Ikai I, Arii S, Kojiro M, Ichida T, Makuuchi M, Matsuyama Y, et al. Reevaluation of prognostic factors for survival after liver resection in patients with hepatocellular carcinoma in a Japanese nationwide survey. Cancer. 2004; 101: 796–802.
  77. Bruix J, Cheng AL, Meinhardt G, Nakajima K, De Sanctis Y, Llovet J. Prognostic factors and predictors of sorafenib benefit in patients with hepatocellular carcinoma: Analysis of two phase III studies. J Hepatol. 2017 Nov; 67: 999–1008.
  78. Tangkijvanich P, Anukulkarnkusol N, Suwangool P, Lertmaharit S, Hanvivatvong O, Kullavanijaya P, et al. Clinical characteristics and prognosis of hepatocellular carcinoma. J Clin Gastroenterol. 2000; 31: 302–308.
  79. Sakata J, Shirai Y, Wakai T, Kaneko K, Nagahashi M, Hatakeyama K. Preoperative predictors of vascular invasion in hepatocellular carcinoma. Eur J Surg Oncol (EJSO). 2008; 34: 900–905.
  80. Tangkijvanich P, Anukulkarnkusol N, Suwangool P, Lertmaharit S, Hanvivatvong O, Kullavanijaya P, et al. Clinical characteristics and prognosis of hepatocellular carcinoma. J Clin Gastroenterol. 2000; 31: 302–308.
  81. Vibert E, Azoulay D, Hoti E, Iacopinelli S, Samuel D, Salloum C, et al. Progression of alphafetoprotein before liver transplantation for hepatocellular carcinoma in cirrhotic patients: A critical factor. Am J Transplant. 2010; 10: 129–137.
  82. Merani S, Majno P, Kneteman NM, Berney T, Morel P, Mentha G, et al. The impact of waiting list alpha-fetoprotein changes on the outcome of liver transplant for hepatocellular carcinoma. J Hepatol. 2011; 55: 814–819.
  83. Sapisochin G, Goldaracena N, Laurence JM, Dib M, Barbas A, Ghanekar A, et al. The extended Toronto criteria for liver transplantation in patients with hepatocellular carcinoma: A prospective validation study. Hepatology. 2016; 64: 2077–2088.
  84. Chan SL, Mo FKF, Johnson PJ, Hui EP, Ma BBY, Ho WM, et al. New utility of an old marker: Serial α-fetoprotein measurement in predicting radiologic response and survival of patients with hepatocellular carcinoma undergoing systemic chemotherapy. J Clin Oncol. 2009; 27: 446–452.
  85. Chen LT, Liu TW, Chao Y, Shiah HS, Chang JY, Juang SH, et al. Alpha- fetoprotein response predicts survival benefits of thalidomide in advanced hepatocellular carcinoma. Aliment Pharmacol Ther. 2005; 22: 217–226.
  86. Shao YY, Lin ZZ, Hsu C, Shen YC, Hsu CH, Cheng AL. Early alpha- fetoprotein response predicts treatment efficacy of antiangiogenic systemic therapy in patients with advanced hepatocellular carcinoma. Cancer. 2010; 116: 4590–4596.
  87. Negri F, Gnetti L, Pedrazzi G, Silini EM, Porta C. Sorafenib and hepatocellular carcinoma: Is alpha-fetoprotein a biomarker predictive of tumor biology and primary resistance? Future Oncol. 2021; 17: 3579–3584.
  88. Zhu AX, Park JO, Ryoo BY, Yen CJ, Poon R, Pastorelli D, et al. Ramucirumab versus placebo as second-line treatment in patients with advanced hepatocellular carcinoma following first-line therapy with sorafenib (REACH): a randomised, double-blind, multicentre, phase 3 trial. Lancet Oncol. 2015; 16: 859–870.
  89. Kelley RK, El-Khoueiry AB, Meyer T, Rimassa L, Merle P, Chan SL, et al. Outcomes by baseline Alpha-Fetoprotein (AFP) levels in the phase III CELESTIAL trial of Cabozantinib (C) versus Placebo (P) in previously treated advanced Hepatocellular Carcinoma (HCC). Annals of Oncology. 2018; 29: 236.
  90. Xu T, Fang Y, Rong A, Wang J. Flexible combination of multiple diagnostic biomarkers to improve diagnostic accuracy. BMC Med Res Methodol. 2015; 15: 94.
  91. Kumar S, Mohan A, Guleria R. Biomarkers in cancer screening, research and detection: present and future: a review. Biomarkers. 2006; 11: 385–405.
  92. Sidransky D. Emerging molecular markers of cancer. Nat Rev Cancer. 2002; 2: 210–219.
  93. Su JQ, Liu JS. Linear combinations of multiple diagnostic markers. J Am Stat Assoc. 1993; 88: 1350–1355.
  94. Sharifianjazi F, Jafari Rad A, Bakhtiari A, Niazvand F, Esmaeilkhanian A, Bazli L, et al. Biosensors and nanotechnology for cancer diagnosis (lung and bronchus, breast, prostate, and colon): a systematic review. Biomed Mater. 2021; 17: 012002.
  95. Teng Y, Li W, Gunasekaran S. Biosensors based on single or multiplebiomarkers for diagnosis of prostate cancer. Biosens Bioelectron X. 2023; 15: 100418.
  96. Jayanthi VSPKSA, Das AB, Saxena U. Recent advances in biosensor development for the detection of cancer biomarkers. Biosens Bioelectron. 2017; 91: 15–23.
  97. Qian L, Li Q, Baryeh K, Qiu W, Li K, Zhang J, et al. Biosensors for early diagnosis of pancreatic cancer: a review. Transl Res. 2019; 213: 67– 89.
  98. Sharifi M, Avadi MR, Attar F, Dashtestani F, Ghorchian H, Rezayat SM, et al. Cancer diagnosis using nanomaterials based electrochemical nanobiosensors. Biosens Bioelectron. 2019; 126: 773–784.
  99. Lakemeyer L, Sander S, Wittau M, Henne-Bruns D, Kornmann M, Lemke J. Diagnostic and prognostic value of CEA and CA19-9 in colorectal cancer. Diseases. 2021; 9: 21.

Jakob Z, Günther SC, Marie-Christine S, Sebesta C (2024) Clinical Relevance of Tumor Markers in Gastrointestinal Cancers: A Critical Analysis and Literature Review. JSM Clin Oncol Res 12(1): 1073.

Received : 29 Jan 2024
Accepted : 06 Feb 2024
Published : 09 Feb 2024
Journals
Annals of Otolaryngology and Rhinology
ISSN : 2379-948X
Launched : 2014
JSM Schizophrenia
Launched : 2016
Journal of Nausea
Launched : 2020
JSM Internal Medicine
Launched : 2016
JSM Hepatitis
Launched : 2016
JSM Oro Facial Surgeries
ISSN : 2578-3211
Launched : 2016
Journal of Human Nutrition and Food Science
ISSN : 2333-6706
Launched : 2013
JSM Regenerative Medicine and Bioengineering
ISSN : 2379-0490
Launched : 2013
JSM Spine
ISSN : 2578-3181
Launched : 2016
Archives of Palliative Care
ISSN : 2573-1165
Launched : 2016
JSM Nutritional Disorders
ISSN : 2578-3203
Launched : 2017
Annals of Neurodegenerative Disorders
ISSN : 2476-2032
Launched : 2016
Journal of Fever
ISSN : 2641-7782
Launched : 2017
JSM Bone Marrow Research
ISSN : 2578-3351
Launched : 2016
JSM Mathematics and Statistics
ISSN : 2578-3173
Launched : 2014
Journal of Autoimmunity and Research
ISSN : 2573-1173
Launched : 2014
JSM Arthritis
ISSN : 2475-9155
Launched : 2016
JSM Head and Neck Cancer-Cases and Reviews
ISSN : 2573-1610
Launched : 2016
JSM General Surgery Cases and Images
ISSN : 2573-1564
Launched : 2016
JSM Anatomy and Physiology
ISSN : 2573-1262
Launched : 2016
JSM Dental Surgery
ISSN : 2573-1548
Launched : 2016
Annals of Emergency Surgery
ISSN : 2573-1017
Launched : 2016
Annals of Mens Health and Wellness
ISSN : 2641-7707
Launched : 2017
Journal of Preventive Medicine and Health Care
ISSN : 2576-0084
Launched : 2018
Journal of Chronic Diseases and Management
ISSN : 2573-1300
Launched : 2016
Annals of Vaccines and Immunization
ISSN : 2378-9379
Launched : 2014
JSM Heart Surgery Cases and Images
ISSN : 2578-3157
Launched : 2016
Annals of Reproductive Medicine and Treatment
ISSN : 2573-1092
Launched : 2016
JSM Brain Science
ISSN : 2573-1289
Launched : 2016
JSM Biomarkers
ISSN : 2578-3815
Launched : 2014
JSM Biology
ISSN : 2475-9392
Launched : 2016
Archives of Stem Cell and Research
ISSN : 2578-3580
Launched : 2014
Annals of Clinical and Medical Microbiology
ISSN : 2578-3629
Launched : 2014
JSM Pediatric Surgery
ISSN : 2578-3149
Launched : 2017
Journal of Memory Disorder and Rehabilitation
ISSN : 2578-319X
Launched : 2016
JSM Tropical Medicine and Research
ISSN : 2578-3165
Launched : 2016
JSM Head and Face Medicine
ISSN : 2578-3793
Launched : 2016
JSM Cardiothoracic Surgery
ISSN : 2573-1297
Launched : 2016
JSM Bone and Joint Diseases
ISSN : 2578-3351
Launched : 2017
JSM Bioavailability and Bioequivalence
ISSN : 2641-7812
Launched : 2017
JSM Atherosclerosis
ISSN : 2573-1270
Launched : 2016
Journal of Genitourinary Disorders
ISSN : 2641-7790
Launched : 2017
Journal of Fractures and Sprains
ISSN : 2578-3831
Launched : 2016
Journal of Autism and Epilepsy
ISSN : 2641-7774
Launched : 2016
Annals of Marine Biology and Research
ISSN : 2573-105X
Launched : 2014
JSM Health Education & Primary Health Care
ISSN : 2578-3777
Launched : 2016
JSM Communication Disorders
ISSN : 2578-3807
Launched : 2016
Annals of Musculoskeletal Disorders
ISSN : 2578-3599
Launched : 2016
Annals of Virology and Research
ISSN : 2573-1122
Launched : 2014
JSM Renal Medicine
ISSN : 2573-1637
Launched : 2016
Journal of Muscle Health
ISSN : 2578-3823
Launched : 2016
JSM Genetics and Genomics
ISSN : 2334-1823
Launched : 2013
JSM Anxiety and Depression
ISSN : 2475-9139
Launched : 2016
Clinical Journal of Heart Diseases
ISSN : 2641-7766
Launched : 2016
Annals of Medicinal Chemistry and Research
ISSN : 2378-9336
Launched : 2014
JSM Pain and Management
ISSN : 2578-3378
Launched : 2016
JSM Women's Health
ISSN : 2578-3696
Launched : 2016
Clinical Research in HIV or AIDS
ISSN : 2374-0094
Launched : 2013
Journal of Endocrinology, Diabetes and Obesity
ISSN : 2333-6692
Launched : 2013
Journal of Substance Abuse and Alcoholism
ISSN : 2373-9363
Launched : 2013
JSM Neurosurgery and Spine
ISSN : 2373-9479
Launched : 2013
Journal of Liver and Clinical Research
ISSN : 2379-0830
Launched : 2014
Journal of Drug Design and Research
ISSN : 2379-089X
Launched : 2014
JSM Bioinformatics, Genomics and Proteomics
ISSN : 2576-1102
Launched : 2014
JSM Chemistry
ISSN : 2334-1831
Launched : 2013
Journal of Trauma and Care
ISSN : 2573-1246
Launched : 2014
JSM Surgical Oncology and Research
ISSN : 2578-3688
Launched : 2016
Annals of Food Processing and Preservation
ISSN : 2573-1033
Launched : 2016
Journal of Radiology and Radiation Therapy
ISSN : 2333-7095
Launched : 2013
JSM Physical Medicine and Rehabilitation
ISSN : 2578-3572
Launched : 2016
Annals of Clinical Pathology
ISSN : 2373-9282
Launched : 2013
Annals of Cardiovascular Diseases
ISSN : 2641-7731
Launched : 2016
Journal of Behavior
ISSN : 2576-0076
Launched : 2016
Annals of Clinical and Experimental Metabolism
ISSN : 2572-2492
Launched : 2016
Clinical Research in Infectious Diseases
ISSN : 2379-0636
Launched : 2013
JSM Microbiology
ISSN : 2333-6455
Launched : 2013
Journal of Urology and Research
ISSN : 2379-951X
Launched : 2014
Journal of Family Medicine and Community Health
ISSN : 2379-0547
Launched : 2013
Annals of Pregnancy and Care
ISSN : 2578-336X
Launched : 2017
JSM Cell and Developmental Biology
ISSN : 2379-061X
Launched : 2013
Annals of Aquaculture and Research
ISSN : 2379-0881
Launched : 2014
Clinical Research in Pulmonology
ISSN : 2333-6625
Launched : 2013
Journal of Immunology and Clinical Research
ISSN : 2333-6714
Launched : 2013
Annals of Forensic Research and Analysis
ISSN : 2378-9476
Launched : 2014
JSM Biochemistry and Molecular Biology
ISSN : 2333-7109
Launched : 2013
Annals of Breast Cancer Research
ISSN : 2641-7685
Launched : 2016
Annals of Gerontology and Geriatric Research
ISSN : 2378-9409
Launched : 2014
Journal of Sleep Medicine and Disorders
ISSN : 2379-0822
Launched : 2014
JSM Burns and Trauma
ISSN : 2475-9406
Launched : 2016
Chemical Engineering and Process Techniques
ISSN : 2333-6633
Launched : 2013
Annals of Clinical Cytology and Pathology
ISSN : 2475-9430
Launched : 2014
JSM Allergy and Asthma
ISSN : 2573-1254
Launched : 2016
Journal of Neurological Disorders and Stroke
ISSN : 2334-2307
Launched : 2013
Annals of Sports Medicine and Research
ISSN : 2379-0571
Launched : 2014
JSM Sexual Medicine
ISSN : 2578-3718
Launched : 2016
Annals of Vascular Medicine and Research
ISSN : 2378-9344
Launched : 2014
JSM Biotechnology and Biomedical Engineering
ISSN : 2333-7117
Launched : 2013
Journal of Hematology and Transfusion
ISSN : 2333-6684
Launched : 2013
JSM Environmental Science and Ecology
ISSN : 2333-7141
Launched : 2013
Journal of Cardiology and Clinical Research
ISSN : 2333-6676
Launched : 2013
JSM Nanotechnology and Nanomedicine
ISSN : 2334-1815
Launched : 2013
Journal of Ear, Nose and Throat Disorders
ISSN : 2475-9473
Launched : 2016
JSM Ophthalmology
ISSN : 2333-6447
Launched : 2013
Journal of Pharmacology and Clinical Toxicology
ISSN : 2333-7079
Launched : 2013
Annals of Psychiatry and Mental Health
ISSN : 2374-0124
Launched : 2013
Medical Journal of Obstetrics and Gynecology
ISSN : 2333-6439
Launched : 2013
Annals of Pediatrics and Child Health
ISSN : 2373-9312
Launched : 2013
JSM Clinical Pharmaceutics
ISSN : 2379-9498
Launched : 2014
JSM Foot and Ankle
ISSN : 2475-9112
Launched : 2016
JSM Alzheimer's Disease and Related Dementia
ISSN : 2378-9565
Launched : 2014
Journal of Addiction Medicine and Therapy
ISSN : 2333-665X
Launched : 2013
Journal of Veterinary Medicine and Research
ISSN : 2378-931X
Launched : 2013
Annals of Public Health and Research
ISSN : 2378-9328
Launched : 2014
Annals of Orthopedics and Rheumatology
ISSN : 2373-9290
Launched : 2013
Journal of Clinical Nephrology and Research
ISSN : 2379-0652
Launched : 2014
Annals of Community Medicine and Practice
ISSN : 2475-9465
Launched : 2014
Annals of Biometrics and Biostatistics
ISSN : 2374-0116
Launched : 2013
JSM Clinical Case Reports
ISSN : 2373-9819
Launched : 2013
Journal of Cancer Biology and Research
ISSN : 2373-9436
Launched : 2013
Journal of Surgery and Transplantation Science
ISSN : 2379-0911
Launched : 2013
Journal of Dermatology and Clinical Research
ISSN : 2373-9371
Launched : 2013
JSM Gastroenterology and Hepatology
ISSN : 2373-9487
Launched : 2013
Annals of Nursing and Practice
ISSN : 2379-9501
Launched : 2014
JSM Dentistry
ISSN : 2333-7133
Launched : 2013
Author Information X