Laboratory Evaluation of Tumor Biomarkers


Introduction

Tumor biomarkers are substances synthesized by cancer cells or other body cells in response to cancer and released into the circulation.[1] Tumor biomarkers vary widely in structure and may be simple molecules, such as catecholamines; well-characterized proteins, such as hormones, enzymes, or gene products; or more heterogeneous glycoproteins or mucins, such as carbohydrate antigen 125 (CA 125), which may be quantified by the antibodies used to measure them. Several important tumor biomarkers such as alpha-fetoprotein (AFP), carcinoembryonic antigen (CEA), and human chorionic gonadotropin (hCG) are oncofetal antigens present in the normal fetus, expressed in minute concentrations by healthy tissues, but expressed at high concentrations by some malignancies.[2]

Assays of tumor biomarkers are employed in various clinical settings and are an integral part of many cancer diagnoses and management plans. Using various techniques, these biomarkers may be assayed in selected body fluids such as blood, urine, and pleural or peritoneal effusions. Assays of tumor biomarkers may aid in the screening and early diagnosis of malignancy, guide treatment decisions, monitor treatment response, assess disease progression, or detect cancer recurrence.[3] 

However, assays of tumor biomarkers have limitations and should not be used as standalone diagnostic tools.[4] The results of tumor biomarker assays are most effective when interpreted with clinical information, imaging studies, and pathological tissue examination to ensure a comprehensive assessment and facilitate an accurate diagnosis.

The ideal tumor biomarker would be an inherently stable molecule with high specificity, sensitivity, accuracy, and reproducibility rates, offering cost-effective screening, diagnosis, and prognostic indication. However, no clinically employed tumor biomarker possesses all these characteristics. Most tumor markers have limitations in specificity, sensitivity, or clinical utility, making it essential to use them in conjunction with other diagnostic tools for comprehensive patient evaluation and management.[1][3][5] 

Etiology and Epidemiology

Cancer is not one disease but a group of diseases characterized by dysregulated cellular growth. Malignant neoplasms have the ability to infiltrate, invade, and destroy surrounding tissues.[6] Cancers are caused by genetic mutations that may be inherited or acquired through exposure to environmental carcinogens.[7] Carcinogens like tobacco smoke, asbestos, ionizing and ultraviolet radiation, and infections are associated with malignancy development.[8] 

Cancer is one of the leading causes of death worldwide, accounting for 10 million deaths yearly.[9] On average, 22.6% of women and 18.6% of men risk developing cancer before the age of 75 years.[10] In younger patients, the most commonly encountered malignancies are hematological. However, in older populations, breast, prostate, lung, and colorectal cancers are the most common; these four malignancies comprise more than half of the cancer diagnoses made worldwide. Cancer incidence is increasing globally due to an aging population, lifestyle changes, and environmental pollution.

Bence Jones protein was the first tumor biomarker described in the literature.[11] Since then, many protein- and hormone-based tumor biomarkers have been described and introduced into clinical practice. More recently, proteomics and genomics technology have permitted the assay of various genetic and molecular tumor biomarkers using microarray techniques.

Specimen Requirements and Procedure

The National Academy of Clinical Biochemistry (NCAB) has established preanalytical quality guidelines for tumor biomarkers.[12] Assays of serum should be collected in red-top containers. Other body fluids to be assayed should be collected in fluid-specific containers.[13] Chromosomal assessment of bone marrow requires 2 to 3 mL of bone marrow from the first pull of the repositioned needle during marrow extraction.[14] Whole blood is required for microarray techniques.[15] Immunohistochemistry staining requires approximately a 1-mL tissue volume; the sample should be deparaffinized and rehydrated before staining.[16]

It is preferable to assay all samples immediately. Tissue or bone marrow samples for chromosomal assessment, fluorescent in situ hybridization, or microarray should not be frozen. Salivary contamination may cause falsely increased concentrations of CEA and carbohydrate antigen 19-9.[17] Specimens can be collected at any time of the day; no diurnal variation has been documented. Specimens should always be collected before any invasive procedures since tissue trauma may cause a transient release of tumor markers into the circulation. For example, prostate-specific antigen (PSA) levels increase following urinary catheterization and prostate biopsy, and CEA levels increase after colonoscopy. Tumor biomarker assays should be ideally repeated after 2 to 3 weeks for additional evidence.[18]

The commonly measured tumor markers are generally stable. However, serum or plasma should be separated from the clot and stored at 4 °C in the short term or below -30 °C as soon as possible; relevant guidelines should be followed if available. For prolonged storage, specimens should be stored at -70 °C.[19] Heat treatments should be avoided, such as those used to deplete serum complement components or inactivate the human immunodeficiency virus. Avoidance of heat treatments is particularly true for PSA and hCG, which may dissociate into its free α- and β-subunits at increased temperatures.[20]

Diagnostic Tests

Many malignancies have one or more tumor biomarkers that are routinely measured in the course of diagnosing, managing, and monitoring for recurrence of the neoplasm. Some of these malignancies and their respective biomarkers are listed in Table 1. Malignancies and Related Tumor Biomarkers.

Table 1. Malignancies and Related Tumor Biomarkers.

 Malignancy   Related tumor markers 

 Bronchogenic: 

 Small cell carcinoma 

 Adenocarcinoma 

 Squamous cell carcinoma

 

 Neuron-specific enolase (NSE), Pro-gastrin–releasing peptide (pro-GRP) 

 Carcinoembryonic antigen (CEA)

 Squamous cell carcinoma antigen (SCC), Cytokeratin 19 fragment

 Ovarian:

 Epithelial 

 Mucinous 

 Nonepithelial 

 

 Carbohydrate antigen 125 (CA 125)

 CEA

 Inhibin A and B 

 Colorectal Adenocarcinoma

 CEA

 Carbohydrate antigen 19-9 (CA 19-9)

 Tissue plasminogen activator (TPA)

 Hepatocellular Carcinoma  Alpha-fetoprotein (AFP)
 Pancreatic Adenocarcinoma

 CA 19-9

 CEA

 Prostate Adenocarcinoma

 Prostate-specific antigen (PSA)

 Prostatic acid phosphatase (PAP)

 Germ cell tumors 

 Human chorionic gonadotropin (HCG)

 AFP

 Lactate dehydrogenase

 Placental alkaline phosphatase 

 Breast 

 Carbohydrate antigen 15-3 (CA 15-3)

 Carbohydrate antigen 27.29 (CA 27.29)

 Estrogen and progesterone receptors (ER, PR)

 Human epidermal growth factor receptor 2 (Her2)

 Urokinase plasminogen activator

 Plasminogen activator inhibitor

Testing Procedures

Various assays can be employed when assessing tumor biomarkers. The most commonly utilized testing procedures include:

  • Enzyme assays: Most enzymatic tumor biomarkers, except PSA, are quantified using an enzyme activity assay; enzymatic activity is used to determine the amount of biomarkers in the tested sample.[1] Enzyme activity assays are performed by adding excess substrate and cofactors to the prepared sample, noting their conversion rate to the product. Alternatively, kinetic enzyme assays may be used, which measure substrate at specific, predetermined intervals. 
  • Immunoassays: Immunoassays are based on the principles of the antigen-antibody reaction; when measuring tumor biomarkers, the biomarker serves as the "antigen," and antigen-specific antibodies are used. Commonly employed immunoassay methods are enzyme-linked immunosorbent assay (ELISA), electrochemiluminescence immunoassay, and immunohistochemistry. Many tumor biomarkers are quantified using immunoassay techniques, including AFP, CEA, hCG, prolactin, calcitonin, and carbohydrate antigens.[21]
  • High-performance liquid chromatography (HPLC): HPLC is primarily employed when identifying catecholamines and their metabolites in plasma and urine. HPLC runs analytes over a column and separates them based on their physical properties.[22] 
  • Immunohistochemistry: Immunohistochemistry is a type of immunoassay employed when tumor biomarkers must be detected in solid tissue samples obtained via biopsy. A thin tissue slice is placed on a glass slide, antibodies are added against the specific antigen, and colorimetric secondary antibodies are used to detect antigen-antibody binding. Immunohistochemistry is utilized in the detection of estrogen and progesterone receptors and Her2.[23] 
  • Fluorescent in situ hybridization (FISH): FISH identifies specific genetic mutations in tumor cells.[24] This technique uses fluorescent-labeled DNA probes, which hybridize to specific target sequences in cells, allowing their visualization under a fluorescence microscope. APC (adenomatous polyposis coli) and ras mutations and Her2 overexpression are identified using FISH.[25]
  • Polymerase chain reaction (PCR): PCR is a technique that amplifies a specific DNA segment by cycling through denaturation, annealing of primers, and extension using a thermostable DNA polymerase. PCR is used to detect the bcr-abl1 fusion present in many forms of chronic myeloid leukemia. PCR is also used to detect microsatellite instability or mutations in genes like K-ras, N-ras, and BRAF, which have prognostic and predictive implications in colorectal cancer.[26] PCR is utilized to detect HER2 gene amplification, which helps identify patients who may benefit from HER2-targeted therapy.
  • Microarrays: Microarrays permit the identification and evaluation of multiple genetic mutations or overexpression simultaneously using a solid two-dimensional support material such as a silicon chip with multiple spots containing sequences from thousands of genes. Fluorescent-labeled complementary DNA from the specimen can hybridize, and the signal is measured. Microarrays have many applications; in oncology, they are used for genetic profiling in ovarian and colorectal cancer, genetically typing leukemias, and identifying the tumor of origin in metastatic lesions.[27]

Interfering Factors

Disadvantages of Tumor Biomarkers 

Variations in sample collection, handling, storage, and assay techniques can alter the biomarker profile in a given sample. Standardization of the preanalytical and analytical variables may mitigate these variations. Very low concentrations of tumor biomarkers are frequently encountered in early-stage tumors; a highly sensitive assay is required.[28] Tumor biomarkers also have inherent drawbacks and disadvantages, including:

  • Lack of specificity: Some tumor biomarkers are produced by normal and cancerous cells and are elevated in many noncancerous conditions. This lack of specificity can lead to false-positive results, potentially leading to unnecessary diagnostic procedures or treatment interventions.[2]
  • Lack of sensitivity: Tumor biomarkers may not be elevated in all individuals with a specific cancer, particularly in the early stages of the disease. This lack of sensitivity can result in false-negative results, where biomarker levels are within normal ranges despite the presence of cancer. Such a scenario may delay the diagnosis and lead to missed opportunities for early intervention and treatment.[29]
  • Biological variability: Individual patients can exhibit biological variability in tumor biomarker levels, making it challenging to establish universal reference ranges or cut-off values for diagnosis or monitoring.[30] Biological factors such as age, gender, genetics, and comorbidities can influence biomarker levels, leading to variations in results among individuals.
  • Analytical variability: Variations in assay platforms, reagents, and laboratory techniques can contribute to analytical variability in tumor marker measurements. Inconsistent methods and lack of standardization can impact the accuracy and reliability of results, hindering data comparison across different laboratories or over time.[31]
  • Limited diagnostic utility: Tumor biomarker assays are unsuitable as standalone diagnostic tools for cancer and should be used in conjunction with other diagnostic methods, such as imaging studies, biopsies, or clinical evaluations, to establish a comprehensive diagnosis.[2] Relying solely on tumor biomarker assays may lead to incomplete or inaccurate diagnostic conclusions.

Commonly Encountered Interferences

  • High-dose hook effect: This effect is characterized by falsely low values at high tumor biomarker concentration and is commonly seen in patients for whom the assay is being performed for the first time.[32] The high-dose hook effect can be avoided by using solid-phase antibodies of higher binding capacity, performing the assay in two sample dilutions, and implementing proper wash steps.
  • Specimen carryover: This interference is most commonly encountered when dealing with high-concentration markers in the assay.
  • Interferences from heterophilic or human antimouse antibodies: Samples from patients who have undergone monoclonal antibody therapy or have circulating anti-animal antibodies may return falsely high or low values.[33] Identifying the presence of interfering antibodies requires a high degree of clinical suspicion that a tumor marker result is incorrect; this clinical suspicion may be strengthened if pertinent clinical information is available. Once suspected, potential interference can be investigated by testing the specimen at various dilutions, retesting after treatment with a commercially available blocking agent, adding additional nonimmune mouse serum to the reaction mixture and reassaying, or by reassaying the specimen using a different method provided by another manufacturer, preferably using a different methodology.[34] Caution should be applied when interpreting these results.
  • Pharmaceutical interference: Anticoagulants such as ethylenediaminetetraacetic acid (EDTA) might interfere with some assays.[35]

Results, Reporting, and Critical Findings

The reported results should include reference intervals specific to the employed method and derived from an appropriate healthy population.[36] If possible, the technique used for the assay should be mentioned when reporting results. If there has been a method or technique change, the laboratory should indicate whether it will likely influence the interpretation of the trend in results. There should be a defined protocol if methods are changed, and the likely effect should be communicated to clinical users before the change.[3] Managing the change may necessitate analyzing the previous specimen by the new method or requesting another specimen to reestablish the baseline or confirm the trend in biomarker concentrations.[37]

Instead of interpreting a single value, observing the overall trend of the biomarker concentration resulting from interval testing over time is more likely to provide valuable insight into the status of the disease. Graphical reporting can offer a clear and concise way to interpret the trend in biomarker concentrations over time.[1] Recording brief clinical information alongside the laboratory data enhances result interpretation. Recommendations on the need for confirmatory specimens and the next assay interval can also be included.

Reporting critical increases in tumor biomarker concentrations and accounting for the analytical performance of the test, biological variations, and individual reference intervals contributes to an earlier diagnosis of relapse. The percentage increase or decrease that constitutes a significant change should be defined, account for analytical and biological variation, delineate the expected rate of change in benign and malignant conditions and report the time between samples.[38] For tumor biomarkers, differences in the magnitudes of their biological variation contribute significantly to these percentages.[39]

The half-life of the tumor biomarker must be considered when interpreting test results. Before surgical intervention, use the known half-life of the biomarker to estimate the time required for the level to decline to a normal or undetectable level.[40] If the quantitative decline of a tumor biomarker will be used to determine the likelihood of complete tumor resection, biomarkers should not be measured until at least 2 weeks and ideally 4 weeks postoperatively.[1] The rate of decline may be affected by underlying comorbidities such as renal or hepatic dysfunction.[41] For example, serum CEA often remains inappropriately elevated in patients with underlying hepatic dysfunction due to inefficient hepatic metabolism of the serum biomarker.[42] Persistently elevated serum β-microglobulin is frequently noted in patients with acute and chronic renal disease; even the small-sized β-microglobulin molecule has difficulty passing through the injured glomerular apparatus.[43]

If appropriate to the specific neoplasm, clinicians should consider ordering a panel of tumor biomarkers to increase the diagnostic sensitivity and specificity of the test.[1] Many malignancies have a heterogeneous cellular composition and express more than one tumor biomarker; measurement of multiple biomarkers is frequently required to achieve a >90% detection sensitivity.[13]

Clinical Significance

Tumor biomarkers each possess some degree of clinical utility and correlate to one or several specific malignancies. Many tumor biomarkers are expressed to some degree by normal, healthy cells or tissues, and levels of circulating biomarkers can be affected by benign conditions. The sensitivity and specificity of each assay must be considered within the context of the clinical condition of the patient.[30] 

There are many national and international guidelines on the selection and utilization of tumor biomarkers. The National Academy of Clinical Biochemistry, the European Group on Tumor Markers, the American Cancer Society, the National Comprehensive Cancer Network, and the National Institute for Health and Care Excellence have established recommendations regarding the use of tumor biomarkers based on the level of available evidence. 

The clinical significance of some commonly measured tumor biomarkers is described in Table 2. The Clinical Significance of Selected Tumor Biomarkers.

Table 2. The Clinical Significance of Selected Tumor Biomarkers.

Tumor biomarker  Family  Chemistry  Clinical Significance  Limitation
Alpha-fetoprotein (AFP) Oncofetal antigen Glycoprotein synthesized from the yolk sac and embryonic liver.  Diagnosis and monitoring of hepatocellular carcinoma, hepatoblastoma, and germ cell tumors. Prognostic marker of germ cell tumor.[3] Elevated in pregnancy, neonates, benign liver diseases, and diseases of the gastrointestinal tract.[44]
Carcinoembryonic antigen (CEA) Oncofetal antigen Glycoprotein isolated from fetal gastrointestinal tissue.  Monitoring response to therapy and relapse of colorectal adenocarcinomas.[45] Serum levels in early-stage and poorly differentiated cancer are low and elevated in benign renal, liver, and lung diseases. Not specific to colorectal malignancy.[45] 
Alkaline phosphatase (ALP) Enzyme Enzymes of bone, placenta, small bowel, and biliary tract. Isoenzymes are more specific. Elevated in osteosarcoma, cholangiocarcinoma, and bony metastases.[46] Serum levels are elevated in normal pregnancy and benign diseases of bone, small bowel, and the heaptobiliary system.[47]
Lactate dehydrogenase (LDH)  Enzyme Enzyme found in almost all body cells that interconverts pyruvate and lactate.  Elevated in almost all malignancies due to its ubiquitous nature. [48] Elevated in many anemias and any disease characterized by cellular destruction.

Prostatic acid phosphatase (PAP) 

Enzyme

 Glycoprotein dimer  Monitoring response to therapy and relapse of prostatic adenocarcinoma. [49] High serum levels encountered in some lysosomal storage disorders and many benign prostatic diseases.
Neuron specific enolase (NSE) Enzyme Dimer of the enzyme enolase, synthesized by neuroendocrine cells. Elevated in neuroblastoma, small cell lung cancer, and pancreatic adenocarcinoma.  Delays in the assay should be avoided.[50]
Human chorionic gonadotropin (hCG) Hormone Glycoprotein hormone synthesized by placental syncytiotrophoblasts. Diagnosis, prognosis, and monitoring treatment response of gestational trophoblastic tumor and germ cell tumors.[3] Elevated in normal pregnancy.[51]
 Prolactin  Hormone  Anterior pituitary hormone   Pituitary adenocarcinoma Diurnal variation is seen. Seruum levels may be elevated due to benign pituitary prolactinomas and in response to many medications.[52]
 Calcitonin  Hormone Mucin glycoprotein secreted by thyroid parafollicular C cells. Diagnosis and monitoring of medullary thyroid carcinoma. Falsely elevated in Zollinger-Ellison syndrome, pernicious anemia, and chronic renal disease.[53]
Catecholamines and metanephrines  Hormone Biogenic amines produced by the adrenal gland and sympathetic nervous system. Diagnosis and monitoring of neuroblastoma, pheochromocytoma, and paragangliomas.[54]   Serum levels may be elevated in response to many medications and normal diurnal variation is seen.
Serotonin Hormone Biogenic amine Diagnosis and monitoring of carcinoid tumors.[55]  Levels may be elevated after consuming meat and fruits.
Prostate-specific antigen (PSA) Protein Glycoprotein with serine protease activity that circulates free or bound to antichymotrypsin or macroglobulin.  Screening, risk assessment, and monitoring for prostatic cancer.[56] Serum levels may be elevated in many benign prostatic diseases and after manipulation of the male lower genitourinary tract.[57]
Carbohydrate antigen 15-3 (CA 15-3) Protein  Mucin glycoprotein Used in conjunction with CEA for monitoring breast cancer. Used as a marker for treatment response. Elevated in benign and malignant breast, ovarian, and liver disease.[50]
Carbohydrate antigen 19-9 (CA 19-9) Protein Lewis blood grouping glycolipid Increased in pancreatic and hepatobiliary cancer. Monitoring pancreatic cancer following resection. Specimens contaminated with saliva may show high CA 19-9 values. Low or absent levels in patients who are negative for the Lewis blood group.[17]
Carbohydrate antigen 125 (CA 125) Protein  Mucin glycoprotein  Screening and monitoring ovarian epithelial carcinoma. CA 125 is also made by the pleurae, pericardium, and peritoneum and will be elevated in benign diseases affecting those tissues.[58]
β-2 microglobulin  Protein Component of major histocompatibility complex Class I (MHC Class I) Increased in chronic lymphocytic leukemia, multiple myeloma, and B-cell neoplasms.[3] May be elevated in acute and chronic renal disease and active HIV infection.
Thyroglobulin  Protein Glycoprotein dimer  Monitoring differentiated thyroid carcinoma.[59]  Autoantibodies of many thyroid diseases falsely elevated serum thyroglobulin. 
Human epidermal growth factor receptor 2 (Her2) Protein Glycoprotein of tyrosine kinase receptor family activation which causes cell growth and proliferation. HER2 overexpression may be seen in breast, ovarian, and endometrial carcinoma.[60] Varying expressions in different areas of the tumor.
Estrogen and progesterone receptors (ER, PR) Protein Nuclear transcription factor and  steroid receptor  Predicting responsiveness of breast cancer to antihormonal therapies.[60]

Biomarker expression changes over time.

 TP53 Genetic marker  Tumor suppressor gene Most commonly mutated gene in human cancer.[3]   Will be elevated in the presence   of some colon polyps. 
Retinoblastoma gene (RB) Genetic marker  Tumor suppressor gene  Directly or indirectly mutated in almost all human cancers. [61]                        -
 BRCA1 and BRCA2  Genetic marker  Tumor suppressor gene  Mutation predisposes to many cancers in both sexes.[3]                      - 
Adenomatous polyposis coli gene (APC) Genetic marker  Tumor suppressor gene Hereditary nonpolyposis colonic, breast, and esophageal adenocarcinomas.[50] Will be elevated in the presence  of some colon polyps.
 ras Genetic marker Proto-oncogene Mutations in ras are found in most human cancers.[62] Mutations are almost ubiquitous and complex.
 C-myc Genetic marker Proto-oncogene T-cell and B-cell lymphomas, small cell lung cancer. Used to identify a high-risk population.[63] Varying expression is seen in different tumor types.
 bcl-2 Genetic marker  Oncogene promoting cell survival Found in leukemia and lymphoma. Presence indicates resistance to chemotherapy.[64]                     - 

Quality Control and Lab Safety

The testing laboratory is responsible for implementing stringent quality control measures to ensure the accuracy and reliability of the test. Assays should be validated before clinical use to ensure the provision of accurate and relevant reports. Recommended intra-assay and interassay variability are <5% and <10%, respectively. Some newer techniques may perform significantly better but may be less precise.[12]

Aspects of quality control, such as internal and proficiency testing (PT), should be implemented. The quality control specimen should mimic sera, and multiple levels can be used to cover the range of concentration, including the decision limits. It is important to include negative and low-positive controls.[65] The number of internal quality control samples to be run for marker assay validation depends on the frequency of testing. The samples should be checked frequently for assay interferences. During tumor marker assay, calibration and daily maintenance should be conducted before running quality control (QC) samples.[66] 

Immediate and appropriate action should be taken to avoid erroneous reporting when an assay run fails to meet objective criteria for assay acceptance. Criteria for acceptance should be predefined and based on logical criteria such as those of Westgard. The number of IQC specimens included per run should allow the identification of an unacceptable run with a given probability appropriate to the clinical application.[67] Given the long-term monitoring of cancer care, assay stability should be ensured over prolonged periods. Laboratories should have procedures and acceptance criteria for assessing lot-to-lot variation that may adversely affect clinical outcomes.[68]

Quality control (QC) material not provided by the method manufacturer is preferable; kit controls may provide an overly optimistic impression of performance as they are unlikely to be commutable with patient serum. At least one authentic serum matrix control from an independent source should be included in addition to any QC materials provided by the method manufacturer.[66]

PT specimens should be commutable with patient specimens to ensure valid between-method comparisons.[69] Concentrations should assess performance over the working range and should include an assessment of linearity on dilution, baseline security, and stability of results over time. The PT provider is responsible for ensuring that specimens are stable in transit. The target values, usually consensus means for heterogeneous analytes, should be accurate and stable, as demonstrated by assessing their accuracy, stability, and linearity on dilution.[70]

When performing tumor biomarker assays, adhere to standard laboratory safety practices, including personal protective equipment, handling and disposal of biohazardous materials properly, and maintaining a clean work environment. Follow equipment maintenance and calibration protocols, and ensure staff is trained in emergency procedures to promote a safe and efficient laboratory environment.[71]

Enhancing Healthcare Team Outcomes

Tumor biomarker assay requires a multifaceted approach. Laboratory technicians with expertise in running tumor marker assays are essential to ensure accurate testing. Lab professionals' roles include selecting appropriate assays for specific cancer types, establishing appropriate cutoff values, and determining the significance of marker trends over time. Clinicians should have a strategy toward evidence-based practices and the clinical utility of tumor marker assays.

Healthcare providers should uphold ethical principles while discussing test results, potential limitations, and implications for treatment choices. Patient safety is of paramount importance throughout the assay process. Adequate measures should be in place to prevent contamination, ensure specimen integrity, and safeguard patient information. Interprofessional communication and coordination are crucial for seamless care. Collaboration among physicians, pathologists, laboratory technicians, and other healthcare professionals ensures accurate sample collection, timely test results, and effective integration of tumor marker data into patient management plans. This collaborative effort enhances care coordination within a concise framework.


Details

Editor:

Muhammad Zubair

Updated:

10/28/2023 9:35:32 PM

References


[1]

Sharma S. Tumor markers in clinical practice: General principles and guidelines. Indian journal of medical and paediatric oncology : official journal of Indian Society of Medical & Paediatric Oncology. 2009 Jan:30(1):1-8. doi: 10.4103/0971-5851.56328. Epub     [PubMed PMID: 20668599]


[2]

Nagpal M, Singh S, Singh P, Chauhan P, Zaidi MA. Tumor markers: A diagnostic tool. National journal of maxillofacial surgery. 2016 Jan-Jun:7(1):17-20. doi: 10.4103/0975-5950.196135. Epub     [PubMed PMID: 28163473]


[3]

Malati T. Tumour markers: An overview. Indian journal of clinical biochemistry : IJCB. 2007 Sep:22(2):17-31. doi: 10.1007/BF02913308. Epub     [PubMed PMID: 23105677]

Level 3 (low-level) evidence

[4]

Bhatt AN, Mathur R, Farooque A, Verma A, Dwarakanath BS. Cancer biomarkers - current perspectives. The Indian journal of medical research. 2010 Aug:132():129-49     [PubMed PMID: 20716813]

Level 3 (low-level) evidence

[5]

Zygulska AL, Pierzchalski P. Novel Diagnostic Biomarkers in Colorectal Cancer. International journal of molecular sciences. 2022 Jan 13:23(2):. doi: 10.3390/ijms23020852. Epub 2022 Jan 13     [PubMed PMID: 35055034]


[6]

Upadhyay A. Cancer: An unknown territory; rethinking before going ahead. Genes & diseases. 2021 Sep:8(5):655-661. doi: 10.1016/j.gendis.2020.09.002. Epub 2020 Sep 18     [PubMed PMID: 34291136]


[7]

Hamilton JG, Waters EA. How are multifactorial beliefs about the role of genetics and behavior in cancer causation associated with cancer risk cognitions and emotions in the US population? Psycho-oncology. 2018 Feb:27(2):640-647. doi: 10.1002/pon.4563. Epub 2017 Oct 26     [PubMed PMID: 29024169]


[8]

Mbemi A, Khanna S, Njiki S, Yedjou CG, Tchounwou PB. Impact of Gene-Environment Interactions on Cancer Development. International journal of environmental research and public health. 2020 Nov 3:17(21):. doi: 10.3390/ijerph17218089. Epub 2020 Nov 3     [PubMed PMID: 33153024]


[9]

Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A, Bray F. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA: a cancer journal for clinicians. 2021 May:71(3):209-249. doi: 10.3322/caac.21660. Epub 2021 Feb 4     [PubMed PMID: 33538338]


[10]

White MC, Holman DM, Boehm JE, Peipins LA, Grossman M, Henley SJ. Age and cancer risk: a potentially modifiable relationship. American journal of preventive medicine. 2014 Mar:46(3 Suppl 1):S7-15. doi: 10.1016/j.amepre.2013.10.029. Epub     [PubMed PMID: 24512933]


[11]

Sewpersad S, Pillay TS. Historical perspectives in clinical pathology: Bence Jones protein-early urine chemistry and the impact on modern day diagnostics. Journal of clinical pathology. 2021 Apr:74(4):212-215. doi: 10.1136/jclinpath-2020-206675. Epub 2020 May 29     [PubMed PMID: 32471887]

Level 3 (low-level) evidence

[12]

Sturgeon CM, Duffy MJ, Stenman UH, Lilja H, Brünner N, Chan DW, Babaian R, Bast RC Jr, Dowell B, Esteva FJ, Haglund C, Harbeck N, Hayes DF, Holten-Andersen M, Klee GG, Lamerz R, Looijenga LH, Molina R, Nielsen HJ, Rittenhouse H, Semjonow A, Shih IeM, Sibley P, Sölétormos G, Stephan C, Sokoll L, Hoffman BR, Diamandis EP, National Academy of Clinical Biochemistry. National Academy of Clinical Biochemistry laboratory medicine practice guidelines for use of tumor markers in testicular, prostate, colorectal, breast, and ovarian cancers. Clinical chemistry. 2008 Dec:54(12):e11-79. doi: 10.1373/clinchem.2008.105601. Epub     [PubMed PMID: 19042984]

Level 1 (high-level) evidence

[13]

Duffy MJ. Clinical uses of tumor markers: a critical review. Critical reviews in clinical laboratory sciences. 2001 Jun:38(3):225-62     [PubMed PMID: 11451209]


[14]

Quintanilla-Martinez L, Tinguely M, Bonzheim I, Fend F. [Bone marrow biopsy: processing and use of molecular techniques]. Der Pathologe. 2012 Nov:33(6):481-9. doi: 10.1007/s00292-012-1647-z. Epub     [PubMed PMID: 23085692]


[15]

Vartanian K, Slottke R, Johnstone T, Casale A, Planck SR, Choi D, Smith JR, Rosenbaum JT, Harrington CA. Gene expression profiling of whole blood: comparison of target preparation methods for accurate and reproducible microarray analysis. BMC genomics. 2009 Jan 5:10():2. doi: 10.1186/1471-2164-10-2. Epub 2009 Jan 5     [PubMed PMID: 19123946]


[16]

Magaki S, Hojat SA, Wei B, So A, Yong WH. An Introduction to the Performance of Immunohistochemistry. Methods in molecular biology (Clifton, N.J.). 2019:1897():289-298. doi: 10.1007/978-1-4939-8935-5_25. Epub     [PubMed PMID: 30539453]


[17]

Kim S, Park BK, Seo JH, Choi J, Choi JW, Lee CK, Chung JB, Park Y, Kim DW. Carbohydrate antigen 19-9 elevation without evidence of malignant or pancreatobiliary diseases. Scientific reports. 2020 Jun 1:10(1):8820. doi: 10.1038/s41598-020-65720-8. Epub 2020 Jun 1     [PubMed PMID: 32483216]


[18]

van der Kwast TH, Lopes C, Santonja C, Pihl CG, Neetens I, Martikainen P, Di Lollo S, Bubendorf L, Hoedemaeker RF, Members of the pathology committee of the European Randomised Study of Screening for Prostate Cancer. Guidelines for processing and reporting of prostatic needle biopsies. Journal of clinical pathology. 2003 May:56(5):336-40     [PubMed PMID: 12719451]

Level 1 (high-level) evidence

[19]

Sturgeon CM, Hoffman BR, Chan DW, Ch'ng SL, Hammond E, Hayes DF, Liotta LA, Petricoin EF, Schmitt M, Semmes OJ, Söletormos G, van der Merwe E, Diamandis EP, National Academy of Clinical Biochemistry. National Academy of Clinical Biochemistry Laboratory Medicine Practice Guidelines for use of tumor markers in clinical practice: quality requirements. Clinical chemistry. 2008 Aug:54(8):e1-e10. doi: 10.1373/clinchem.2007.094144. Epub 2008 Jul 7     [PubMed PMID: 18606634]

Level 1 (high-level) evidence

[20]

Mosley AK, Brouwer KL. Heat treatment of human serum to inactivate HIV does not alter protein binding of selected drugs. Therapeutic drug monitoring. 1997 Aug:19(4):477-9     [PubMed PMID: 9263392]


[21]

Yin Y, Cao Y, Xu Y, Li G. Colorimetric immunoassay for detection of tumor markers. International journal of molecular sciences. 2010:11(12):5077-94. doi: 10.3390/ijms11125077. Epub 2010 Dec 7     [PubMed PMID: 21614193]


[22]

Hjemdahl P. Catecholamine measurements by high-performance liquid chromatography. The American journal of physiology. 1984 Jul:247(1 Pt 1):E13-20     [PubMed PMID: 6377918]


[23]

Zaha DC. Significance of immunohistochemistry in breast cancer. World journal of clinical oncology. 2014 Aug 10:5(3):382-92. doi: 10.5306/wjco.v5.i3.382. Epub     [PubMed PMID: 25114853]


[24]

Ishida C, Zubair M, Gupta V. Molecular Genetics Testing. StatPearls. 2023 Jan:():     [PubMed PMID: 32809547]


[25]

Signoroni S, Vitellaro M, Sala P, Bertario L. Biomarkers in familial adenomatous polyposis: role and significance. Frontiers in bioscience (Scholar edition). 2010 Jan 1:2(2):413-21     [PubMed PMID: 20036957]


[26]

Raj GV, Moreno JG, Gomella LG. Utilization of polymerase chain reaction technology in the detection of solid tumors. Cancer. 1998 Apr 15:82(8):1419-42     [PubMed PMID: 9554517]


[27]

Virtanen C, Woodgett J. Clinical uses of microarrays in cancer research. Methods in molecular medicine. 2008:141():87-113     [PubMed PMID: 18453086]


[28]

Agrawal L, Engel KB, Greytak SR, Moore HM. Understanding preanalytical variables and their effects on clinical biomarkers of oncology and immunotherapy. Seminars in cancer biology. 2018 Oct:52(Pt 2):26-38. doi: 10.1016/j.semcancer.2017.12.008. Epub 2017 Dec 16     [PubMed PMID: 29258857]

Level 3 (low-level) evidence

[29]

Ankerst DP, Thompson IM. Sensitivity and specificity of prostate-specific antigen for prostate cancer detection with high rates of biopsy verification. Archivio italiano di urologia, andrologia : organo ufficiale [di] Societa italiana di ecografia urologica e nefrologica. 2006 Dec:78(4):125-9     [PubMed PMID: 17269614]


[30]

Coşkun A, Aarsand AK, Sandberg S, Guerra E, Locatelli M, Díaz-Garzón J, Fernandez-Calle P, Ceriotti F, Jonker N, Bartlett WA, Carobene A, European Federation of Clinical Chemistry and Laboratory Medicine Working Group on Biological Variation. Within- and between-subject biological variation data for tumor markers based on the European Biological Variation Study. Clinical chemistry and laboratory medicine. 2022 Mar 28:60(4):543-552. doi: 10.1515/cclm-2021-0283. Epub 2021 May 10     [PubMed PMID: 33964202]


[31]

Sturgeon C. Standardization of tumor markers - priorities identified through external quality assessment. Scandinavian journal of clinical and laboratory investigation. Supplementum. 2016:245():S94-9. doi: 10.1080/00365513.2016.1210334. Epub     [PubMed PMID: 27542005]

Level 2 (mid-level) evidence

[32]

Jassam N, Jones CM, Briscoe T, Horner JH. The hook effect: a need for constant vigilance. Annals of clinical biochemistry. 2006 Jul:43(Pt 4):314-7     [PubMed PMID: 16824284]


[33]

Reinsberg J. Interference by human antibodies with tumor marker assays. Hybridoma. 1995 Apr:14(2):205-8     [PubMed PMID: 7590781]


[34]

Bolstad N, Warren DJ, Nustad K. Heterophilic antibody interference in immunometric assays. Best practice & research. Clinical endocrinology & metabolism. 2013 Oct:27(5):647-61. doi: 10.1016/j.beem.2013.05.011. Epub 2013 Jun 20     [PubMed PMID: 24094636]


[35]

Banfi G, Salvagno GL, Lippi G. The role of ethylenediamine tetraacetic acid (EDTA) as in vitro anticoagulant for diagnostic purposes. Clinical chemistry and laboratory medicine. 2007:45(5):565-76     [PubMed PMID: 17484616]


[36]

Horn PS, Pesce AJ. Reference intervals: an update. Clinica chimica acta; international journal of clinical chemistry. 2003 Aug:334(1-2):5-23     [PubMed PMID: 12867273]


[37]

Hayes DF. Biomarker validation and testing. Molecular oncology. 2015 May:9(5):960-6. doi: 10.1016/j.molonc.2014.10.004. Epub 2014 Oct 18     [PubMed PMID: 25458054]

Level 1 (high-level) evidence

[38]

Duffy MJ. Tumor markers in clinical practice: a review focusing on common solid cancers. Medical principles and practice : international journal of the Kuwait University, Health Science Centre. 2013:22(1):4-11. doi: 10.1159/000338393. Epub 2012 May 15     [PubMed PMID: 22584792]


[39]

Marques-Garcia F, Boned B, González-Lao E, Braga F, Carobene A, Coskun A, Díaz-Garzón J, Fernández-Calle P, Perich MC, Simon M, Jonker N, Aslan B, Bartlett WA, Sandberg S, Aarsand AK, European Federation of Clinical Chemistry and Laboratory Medicine Working Group on Biological Variation and Task Group for the Biological Variation Database. Critical review and meta-analysis of biological variation estimates for tumor markers. Clinical chemistry and laboratory medicine. 2022 Mar 28:60(4):494-504. doi: 10.1515/cclm-2021-0725. Epub 2022 Feb 10     [PubMed PMID: 35143717]

Level 1 (high-level) evidence

[40]

Arlen PM, Bianco F, Dahut WL, D'Amico A, Figg WD, Freedland SJ, Gulley JL, Kantoff PW, Kattan MW, Lee A, Regan MM, Sartor O, Prostate Specific Antigen Working Group. Prostate Specific Antigen Working Group guidelines on prostate specific antigen doubling time. The Journal of urology. 2008 Jun:179(6):2181-5; discussion 2185-6. doi: 10.1016/j.juro.2008.01.099. Epub 2008 Apr 18     [PubMed PMID: 18423743]


[41]

Coppolino G, Bolignano D, Rivoli L, Mazza G, Presta P, Fuiano G. Tumour markers and kidney function: a systematic review. BioMed research international. 2014:2014():647541. doi: 10.1155/2014/647541. Epub 2014 Feb 6     [PubMed PMID: 24689048]

Level 1 (high-level) evidence

[42]

Maestranzi S, Przemioslo R, Mitchell H, Sherwood RA. The effect of benign and malignant liver disease on the tumour markers CA19-9 and CEA. Annals of clinical biochemistry. 1998 Jan:35 ( Pt 1)():99-103     [PubMed PMID: 9463746]


[43]

Al-Taee IK, Al-Safar JJ, Al-Falahi YS, Al-Shamma IA. The Clinical Significance of beta2-microglobulin in End-Stage Renal Disease. Saudi journal of kidney diseases and transplantation : an official publication of the Saudi Center for Organ Transplantation, Saudi Arabia. 2003 Oct-Dec:14(4):492-6     [PubMed PMID: 17657122]


[44]

Wong RJ, Ahmed A, Gish RG. Elevated alpha-fetoprotein: differential diagnosis - hepatocellular carcinoma and other disorders. Clinics in liver disease. 2015 May:19(2):309-23. doi: 10.1016/j.cld.2015.01.005. Epub 2015 Feb 27     [PubMed PMID: 25921665]


[45]

Hammarström S. The carcinoembryonic antigen (CEA) family: structures, suggested functions and expression in normal and malignant tissues. Seminars in cancer biology. 1999 Apr:9(2):67-81     [PubMed PMID: 10202129]


[46]

Narayanan S. Alkaline phosphatase as tumor marker. Annals of clinical and laboratory science. 1983 Mar-Apr:13(2):133-6     [PubMed PMID: 6859804]


[47]

Kim SH, Shin KH, Moon SH, Jang J, Kim HS, Suh JS, Yang WI. Reassessment of alkaline phosphatase as serum tumor marker with high specificity in osteosarcoma. Cancer medicine. 2017 Jun:6(6):1311-1322. doi: 10.1002/cam4.1022. Epub 2017 May 11     [PubMed PMID: 28493412]


[48]

Jurisic V, Radenkovic S, Konjevic G. The Actual Role of LDH as Tumor Marker, Biochemical and Clinical Aspects. Advances in experimental medicine and biology. 2015:867():115-24. doi: 10.1007/978-94-017-7215-0_8. Epub     [PubMed PMID: 26530363]

Level 3 (low-level) evidence

[49]

Kong HY, Byun J. Emerging roles of human prostatic Acid phosphatase. Biomolecules & therapeutics. 2013 Jan:21(1):10-20. doi: 10.4062/biomolther.2012.095. Epub     [PubMed PMID: 24009853]


[50]

Beketic-Oreskovic L, Maric P, Ozretic P, Oreskovic D, Ajdukovic M, Levanat S. Assessing the clinical significance of tumor markers in common neoplasms. Frontiers in bioscience (Elite edition). 2012 Jun 1:4(7):2558-78     [PubMed PMID: 22652661]


[51]

Boime I, Ben-Menahem D. Glycoprotein hormone structure-function and analog design. Recent progress in hormone research. 1999:54():271-88; discussion 288-9     [PubMed PMID: 10548880]


[52]

Majumdar A, Mangal NS. Hyperprolactinemia. Journal of human reproductive sciences. 2013 Jul:6(3):168-75. doi: 10.4103/0974-1208.121400. Epub     [PubMed PMID: 24347930]


[53]

Kiriakopoulos A, Giannakis P, Menenakos E. Calcitonin: current concepts and differential diagnosis. Therapeutic advances in endocrinology and metabolism. 2022:13():20420188221099344. doi: 10.1177/20420188221099344. Epub 2022 May 21     [PubMed PMID: 35614985]

Level 3 (low-level) evidence

[54]

Neumann HPH, Young WF Jr, Eng C. Pheochromocytoma and Paraganglioma. The New England journal of medicine. 2019 Aug 8:381(6):552-565. doi: 10.1056/NEJMra1806651. Epub     [PubMed PMID: 31390501]


[55]

Maroun J, Kocha W, Kvols L, Bjarnason G, Chen E, Germond C, Hanna S, Poitras P, Rayson D, Reid R, Rivera J, Roy A, Shah A, Sideris L, Siu L, Wong R. Guidelines for the diagnosis and management of carcinoid tumours. Part 1: the gastrointestinal tract. A statement from a Canadian National Carcinoid Expert Group. Current oncology (Toronto, Ont.). 2006 Apr:13(2):67-76     [PubMed PMID: 17576444]


[56]

Asif S, Teply BA. Biomarkers for Treatment Response in Advanced Prostate Cancer. Cancers. 2021 Nov 16:13(22):. doi: 10.3390/cancers13225723. Epub 2021 Nov 16     [PubMed PMID: 34830878]


[57]

Chung MS, Lee SH. Current status of active surveillance in prostate cancer. Investigative and clinical urology. 2016 Jan:57(1):14-20. doi: 10.4111/icu.2016.57.1.14. Epub 2016 Jan 11     [PubMed PMID: 26966722]


[58]

Rein BJ, Gupta S, Dada R, Safi J, Michener C, Agarwal A. Potential markers for detection and monitoring of ovarian cancer. Journal of oncology. 2011:2011():475983. doi: 10.1155/2011/475983. Epub 2011 Apr 11     [PubMed PMID: 21577260]


[59]

Prpić M, Franceschi M, Romić M, Jukić T, Kusić Z. THYROGLOBULIN AS A TUMOR MARKER IN DIFFERENTIATED THYROID CANCER - CLINICAL CONSIDERATIONS. Acta clinica Croatica. 2018 Sep:57(3):518-527. doi: 10.20471/acc.2018.57.03.16. Epub     [PubMed PMID: 31168186]


[60]

Onitilo AA, Engel JM, Greenlee RT, Mukesh BN. Breast cancer subtypes based on ER/PR and Her2 expression: comparison of clinicopathologic features and survival. Clinical medicine & research. 2009 Jun:7(1-2):4-13. doi: 10.3121/cmr.2009.825. Epub     [PubMed PMID: 19574486]


[61]

Dimaras H, Corson TW, Cobrinik D, White A, Zhao J, Munier FL, Abramson DH, Shields CL, Chantada GL, Njuguna F, Gallie BL. Retinoblastoma. Nature reviews. Disease primers. 2015 Aug 27:1():15021. doi: 10.1038/nrdp.2015.21. Epub 2015 Aug 27     [PubMed PMID: 27189421]


[62]

Fernández-Medarde A, Santos E. Ras in cancer and developmental diseases. Genes & cancer. 2011 Mar:2(3):344-58. doi: 10.1177/1947601911411084. Epub     [PubMed PMID: 21779504]


[63]

Nguyen L, Papenhausen P, Shao H. The Role of c-MYC in B-Cell Lymphomas: Diagnostic and Molecular Aspects. Genes. 2017 Apr 5:8(4):. doi: 10.3390/genes8040116. Epub 2017 Apr 5     [PubMed PMID: 28379189]


[64]

Thomas S, Quinn BA, Das SK, Dash R, Emdad L, Dasgupta S, Wang XY, Dent P, Reed JC, Pellecchia M, Sarkar D, Fisher PB. Targeting the Bcl-2 family for cancer therapy. Expert opinion on therapeutic targets. 2013 Jan:17(1):61-75. doi: 10.1517/14728222.2013.733001. Epub 2012 Nov 22     [PubMed PMID: 23173842]

Level 3 (low-level) evidence

[65]

Badrick T. Quality leadership and quality control. The Clinical biochemist. Reviews. 2003 Aug:24(3):81-93     [PubMed PMID: 18568046]

Level 2 (mid-level) evidence

[66]

Kinns H, Pitkin S, Housley D, Freedman DB. Internal quality control: best practice. Journal of clinical pathology. 2013 Dec:66(12):1027-32. doi: 10.1136/jclinpath-2013-201661. Epub 2013 Sep 26     [PubMed PMID: 24072731]

Level 2 (mid-level) evidence

[67]

Mrazek C, Lippi G, Keppel MH, Felder TK, Oberkofler H, Haschke-Becher E, Cadamuro J. Errors within the total laboratory testing process, from test selection to medical decision-making - A review of causes, consequences, surveillance and solutions. Biochemia medica. 2020 Jun 15:30(2):020502. doi: 10.11613/BM.2020.020502. Epub     [PubMed PMID: 32550813]


[68]

Abdollahi A, Saffar H, Saffar H. Types and Frequency of Errors during Different Phases of Testing At a Clinical Medical Laboratory of a Teaching Hospital in Tehran, Iran. North American journal of medical sciences. 2014 May:6(5):224-8. doi: 10.4103/1947-2714.132941. Epub     [PubMed PMID: 24926448]


[69]

Vesper HW, Miller WG, Myers GL. Reference materials and commutability. The Clinical biochemist. Reviews. 2007 Nov:28(4):139-47     [PubMed PMID: 18392124]


[70]

Miller WG. The role of proficiency testing in achieving standardization and harmonization between laboratories. Clinical biochemistry. 2009 Mar:42(4-5):232-5. doi: 10.1016/j.clinbiochem.2008.09.004. Epub     [PubMed PMID: 19863911]


[71]

Ionescu G, Neguţ M, Combiescu AA. [Biosafety and biosecurity in the medical laboratory. Update and trends]. Bacteriologia, virusologia, parazitologia, epidemiologia (Bucharest, Romania : 1990). 2007 Jul-Dec:52(3-4):91-9     [PubMed PMID: 19326721]