Tumor markers


Abstract

Background

Tumor markers are substances present in and produced by a tumor or produced by the host in response to a tumor. Measured qualitatively or quantitatively by chemical, immunologic, genomic, or proteomic methods, tumor markers can be used to identify the likely presence of a cancer and/or to differentiate a tumor from normal tissue. Tumor markers can contribute to cancer management as screening tests for malignancy in asymptomatic patients, diagnostic aids, prognostic indicators, therapy predictors, and/or post-treatment monitoring. Reflecting the heterogeneous nature of cancer, tumor markers encompass a variety of tumor-derived or tumor-associated molecular species. Tumor markers can be produced by different tumor types. Few are organ-specific and few are specific for a particular malignancy. Tumor markers range from simple molecules (e.g., catecholamines) through relatively well-characterized proteins (e.g., hormones, enzymes, and gene products) to very large heterogeneous glycoproteins and mucins (e.g., CA125), which may be defined by the antibodies used to measure them, and the growing number of deoxyribonucleic acid (DNA) based markers for gene mutations or amplifications (e.g., HER2, EGFR). Although their structures vary widely, the same general principles apply to all tumor markers currently used in clinical practice.

Content

This chapter provides a brief overview of cancer, highlighting its variability and some of the current clinical challenges this important family of diseases poses to laboratory medicine. These are linked with chronological developments in tumor marker applications, from the early recognition of the importance of Bence Jones proteins in myeloma through to molecular and genetic assessment of mutations in solid tumors. Requirements of the “ideal” tumor marker are considered before reviewing the general principles that guide the effective use of tumor markers (i.e., requirements that must be met in the preanalytical, analytical, and postanalytical phases of laboratory service provision). The tumor markers most frequently used in routine practice are reviewed, and requirements for provision of an optimal tumor marker service are considered.

Tumor markers are substances that may be present in abnormally high concentrations in body fluids or tissue from patients with cancer. Tumor markers can aid cancer management in a number of ways, including screening, diagnosis, prognostic assessment, therapy prediction, and/or post-treatment monitoring. Reflecting the heterogeneous nature of cancer, tumor markers encompass a variety of molecular species, which may be tumor-derived or tumor-associated. Many tumor markers are produced by a variety of different tumors, and few tumor markers are organ-specific or specific for a single type of malignancy.

Tumor markers range from simple molecules (e.g., catecholamines) to relatively well-characterized proteins (e.g., hormones, enzymes, and gene products) to much more heterogeneous glycoproteins and mucins (e.g., CA125), which may be defined by the antibodies used to measure them. Several important tumor markers (e.g., α-fetoprotein [AFP], carcinoembryonic antigen [CEA], and human chorionic gonadotropin [hCG]) are oncofetal antigens, which are present in the fetus in normal pregnancy but are expressed at high concentrations in tissue or body fluids of adults with some cancers. Tumor markers are present in cells, tissues, or body fluids and can be measured either qualitatively or quantitatively using chemical, immunologic, or molecular biological methods. While their structures and properties vary widely, the broad principles underpinning their evidence-based clinical application are common to all tumor markers.

This chapter provides a brief overview of cancer and a history of tumor marker development, followed by a comprehensive review of these broad principles. Clinically relevant tumor markers for a number of malignancies are then discussed in detail, considering current practice and future requirements.

Cancer: An overview

Cancer is the name given to a collection of heterogeneous but related diseases that can start in most parts of the body and that are characterized by uncontrolled division of cells that ultimately may spread into surrounding tissues. It is rarely clear what leads to this autonomous growth in individual patients, with causative factors likely to be multifactorial and dependent on the organ involved. Factors that have been implicated include exposure to carcinogens, which may be physical (e.g., radiation), chemical (e.g., polycyclic hydrocarbons), or biological (e.g., viral). Exposure to such agents may cause cancer by direct genotoxic effects on deoxyribonucleic acid (DNA) (e.g., as with radiation) or by increasing cell proliferation (e.g., by a hormone) or both (e.g., through use of tobacco). Excess weight, physical inactivity, and poor nutrition may also contribute to the development of some cancers.

Genetic predisposition to some cancers, including some familial cancers (e.g., MEN2, BRCA1) is increasingly identifiable due to advances in molecular techniques. However, simple genetic analysis of mutations within cancerous cells may be too simplistic to explain a complex process. Sophisticated interaction and communication of cancer cells with their surrounding cellular microenvironment are likely to be essential for both survival and dissemination of cancer cells. Improved understanding of such interactions is already leading to more finely targeted therapies.

For the present, cancer remains a leading cause of death in the United States, second only to heart disease, although death rates have decreased significantly over the last three decades with a 29% fall from 1991 to 2017, including a 2.2% drop from 2016 to 2017, the largest single-year drop ever recorded by the American Cancer Society (ACS). Such figures suggest that early detection and more effective treatment combined with prevention (e.g., decreased smoking, improved diet) and availability of new therapies could reduce the future mortality rate for cancer. However, the identification of smaller more indolent cancers with newer techniques and screening programs may also be contributing to the perceived increase in survival.

Death rates for individual cancers vary markedly ( Table 33.1 ). On comparing the number of deaths per year with the number of new cases, it is clear that lung and bronchus cancer, pancreatic cancer, and hepatocellular carcinoma (HCC) are among the most lethal (59.3, 81.7, and 70.5%, respectively).

TABLE 33.1
Estimated Number of New Cancer Cases and Deaths in the United States for Some Common Cancers (2020 Figures)
Data for incidence and deaths from the American Cancer Society. Estimated new cases are based on incidence rates (through 2016) reported by the Surveillance, Epidemiology and End Results Program, the National Program of Cancer Registries and the North American Association of Central Cancer Registries. Estimated deaths (through 2017) are based on data collected by the National Center for Health Statistics.
Cancer Incidence Number of Deaths Number of Deaths as a Percentage of Incidence
Bladder 81,400 17,980 22.1
Breast 279,100 42,690 15.3
Cervix 13,800 4,290 31.1
Colorectal (colon, rectum, anorectal) 156,540 54,550 34.8
Gastric 27,600 11,010 39.9
Hepatocellular carcinoma 42,810 30,160 70.5
Lung and bronchus 228,820 135,720 59.3
Melanoma 100,350 6,850 6.8
Non-Hodgkin lymphoma 77,240 19,940 25.8
Ovarian 21,750 13,940 64.1
Pancreatic 57,600 47,050 81.7
Prostate 191,930 33,330 17.4
Testicular 9,610 440 4.6
Thyroid 52,890 2,180 4.1
Unknown 30,270 45,850 Near 100
Note: Lack of specificity in recording the underlying cause of death on death certificates and/or an undercount in the case estimate probably accounts for figures suggesting more deaths than cases.

Long-term decreases in death rates for lung, colorectal, breast, and prostate cancers have been documented, although reductions from 2008 to 2017 slowed for female breast and colorectal cancers, and stopped for prostate cancer. The decline for lung cancer has been maintained, with incidence decreasing by 2 to 3% annually during 2008 through 2013 and then by 4 to 5% from 2016 to 2017. This is reflected in the fall in the estimated number of deaths from lung cancer as a percentage of incidence from 71.8% in 2017 to 59.3% in 2020 and contributes to the overall decline in cancer deaths over recent decades. Nevertheless in 2017 more deaths were caused by lung cancer than by prostate, pancreatic, and colorectal cancers combined.

The figures in Table 33.1 reflect some of the considerable advances in treatment over recent decades. Many patients survive for much longer than previously. Although surgical removal of a primary tumor is the only curative therapy for almost all cancers, new chemotherapeutic and biological therapies that prevent progression are increasingly available ( Table 33.2 ). Mortality for melanoma of the skin has decreased by almost twofold since the Food and Drug Administration approved new therapies for metastatic disease. It has been suggested that some relatively indolent cancers should be considered to be chronic diseases like diabetes, to be controlled rather than cured. Breast and prostate cancers are among the best examples. However, for these and other cancers, distinguishing cancers that are likely to progress rapidly from those that are slow-growing or indolent is critical for treatment selection. This distinction remains a challenge for many cancers, demonstrating both the heterogeneity of different cancers and the need for good understanding of their natural history, including at the molecular level.

TABLE 33.2
Treatment Options for Malignancies
Treatment Relevant Malignancies a
Active surveillance Prostate cancer
Curative surgery (curative only in early-stage disease for most malignancies) Early-stage bladder, breast, cervical, colorectal, gastric, germ cell, GIST, GTD, HCC, liver, lung, melanoma, ovarian, prostate, thyroid, and other cancers
Hepatic resection Advanced colorectal cancer
Liver transplant Advanced hepatocellular carcinoma
Palliative surgery Advanced colorectal cancer
Radiotherapy Bladder, breast, cervical, colorectal, gastric, germ cell, lung, prostate, and thyroid cancers
Brachytherapy Prostate cancer
Radioiodine Thyroid cancer
Chemotherapy Bladder, breast, cervical, colorectal, germ cell, GTD, HCC, lung, ovarian, prostate, and other cancers
Ablative therapy (alcohol, radiofrequency, microwave, chemoembolization) Hepatocellular carcinoma
Endocrine therapy Breast and prostate cancer
Immunotherapy Bladder, breast, colorectal, gastric, GIST, lung, melanoma, and non–small cell lung cancers
Targeted therapy Non–small cell lung cancer
GIST, Gastrointestinal stromal tumors; GTD, gestational trophoblastic disease; HCC, hepatocellular carcinoma.

a Treatment of testicular cancers as for germ cell tumors. Treatment of cancers of unknown primary dependent on immunohistochemistry.

Early detection of malignancy optimizes any opportunities for curative surgery for some in situ cancers. Unfortunately, most cancers do not produce symptoms until tumors are too large to be removed surgically or until cancerous cells have spread to other tissue either by invading local lymph nodes or by distant spread (metastasis) to other organs. Systemic treatments (chemotherapy, endocrine therapy, or immunotherapy) are then usually the only options but are not curative. Any residual viable cancerous cells remaining after treatment may proliferate, develop resistance to further therapy, and ultimately cause the death of the patient.

Historical background

The timeline shown in Fig. 33.1 illustrates how closely introduction of new tumor markers has mirrored developments in analytical techniques and more recently has been influenced by the availability of new therapies, many of which are effective only in subsets of cancer patients. Precipitation of a protein from acidified boiled urine in 1847 heralded the identification of Bence Jones protein, the first tumor marker to be used clinically. Characterized more than a century later by the Nobel Prize–winning studies of Porter, Edelman, and Poulik as the monoclonal light chain of immunoglobulin secreted by tumor plasma cells, measurement of Bence Jones protein (paraproteins or M-protein) still forms the basis of many diagnoses of multiple myeloma (see Chapter 31 ).

FIGURE 33.1, Timeline highlighting some of the major advances in tumor marker testing since the early identification of Bence Jones proteins and their association with hematologic malignancy in the 19th century. CEA , Carcinoembryonic antigen; EGFR , epidermal growth factor receptor; ER , estrogen receptor; hCG , human chorionic gonadotropin; LDH , lactate dehydrogenase; PR , progesterone receptor; PSA , prostate-specific antigen; RIA , radioimmunoassay.

In the first half of the 20th century, the presence or absence of several hormones, enzymes, and isoenzymes and blood group antigens were recognized to be associated with malignancy, but it was not until the development of the technique of radioimmunoassay (RIA) in the 1960s that these observations could be translated into routine clinical practice. The application of RIA to the measurement of previously identified oncofetal antigens, including AFP, hCG, and CEA, facilitated their clinical use. Monoclonal antibody technology subsequently enabled the development of two-site immunoradiometric assays (IRMAs) of complex cancer-associated mucins identified primarily by their reactivity with given monoclonal antibody pairs (e.g., the carbohydrate or cancer-associated antigens CA125 and CA15-3). The radiolabels initially used were soon replaced with less hazardous nonisotopic labels, providing immunoenzymatic assays (IEMAs), immunochemiluminescent assays (ICMAs), and immunofluorescent assays (IFMAs) that were much more readily automated than, and rapidly replaced, assays requiring isotopic labels (see Chapter 26 ). As a consequence, tumor marker measurements, which had previously generally been provided in specialist laboratories, became widely available.

Advances in molecular genetics using monoclonal antibodies and molecular probes to detect chromosome or protein alterations, including the study of oncogenes, suppressor genes, and genes involved in DNA repair, have led to rapid understanding and use of tumor markers at both molecular and cellular levels, particularly in high-risk individuals. Estrogen, progesterone, and epidermal growth factor receptor (EGFR) measurements are now routinely performed for breast cancer patients to enable identification of optimal treatment, while establishing the presence or absence of the breast cancer susceptibility genes BRCA1 and BRCA 2 provides additional prognostic information. Similarly, the presence of EGFR mutations and anaplastic lymphoma kinase (ALK) translocations can identify subsets of lung cancer patients who are likely to benefit from targeted molecular therapies.

Mass spectrometry is also increasingly used both as a discovery and a diagnostic tool, albeit with limited success thus far in translating new tumor marker tests into clinical practice (see Chapter 24 ). These developments, for which sophisticated bioinformatics support (e.g., neural networks, support vector machines, and other algorithms [see Chapter 13] ) is essential, are encouraging the use of multiparametric analysis for cancer diagnosis, prognosis, and therapy prediction. The last is becoming increasingly important, with the advent of new and expensive immunotherapies that are effective only in relatively small subsets of patients whose tumors fulfill certain well-defined molecular criteria.

General principles guiding the use of all tumor markers

Tumor markers are surrogate indicators that can help to increase or decrease the clinical suspicion that a future clinically important event, such as the development of a new or secondary cancer, recurrence, progression, or death will or will not occur, and/or that a specific treatment will reduce that risk. Tumor markers can help make or confirm a cancer diagnosis, monitor treatment effectiveness and the course of disease, estimate prognosis, and/or predict whether a specific therapy is likely to be successful ( Table 33.3 ). Their measurement should permit more efficient application of therapies by ensuring that these are applied only to those patients most likely to benefit and reducing exposure to unnecessary toxicity for those patients unlikely to benefit. , Tumor markers should be measured only after careful consideration of whether the result is likely to provide information that may improve outcome for the individual patient , or if required as part of a clinical trial.

TABLE 33.3
Current Clinical Applications of Tumor Markers
Clinical Application Requirements Examples
Screening for cancer An acceptable test for a disease that poses an important health problem, for which the natural history is well understood, the test identifies treatable early-stage cancers, and early intervention with effective treatment improves outcome. High clinical sensitivity (few false negatives) and specificity (few false positives) are essential prerequisites because the population screened is asymptomatic. Population screening: Fecal occult blood testing for colorectal cancer. ,
Screening of high-risk groups: Serum hCG testing for choriocarcinoma in women who have had a molar pregnancy.
Diagnosing cancer High clinical sensitivity and specificity as for screening. Most serum tumor markers are not cancer-specific and/or organ-specific and are raised in different cancers and/or in nonmalignant disease, severely limiting their use in diagnosis. As a diagnostic aid in high-risk groups: Serum AFP testing as an adjunct to ultrasound for hepatocellular carcinoma (HCC) in patients with cirrhosis who are at high risk of developing HCC.
Differential diagnosis: CA125 contributes (with menopausal status and ultrasound findings) to calculate a “risk of malignancy index,” which is used to differentiate patients with benign and malignant pelvic masses. ,
Assessing prognosis A test that can provide a probability estimate of outcome (risk of relapse or disease progression) and/or differentiate indolent from aggressive disease for a heterogeneous population of patients, thereby influencing treatment decisions. Serum AFP, hCG, and lactate dehydrogenase (LDH) measurements are mandatory for determining prognosis and selection of chemotherapy in patients with metastatic nonseminomatous germ cell tumors.
Microsatellite instability (MSI) occurs when germline alleles of microsatellites (short stretches of DNA that are repeated at multiple locations throughout the genome) lose or gain a repeat unit due to failure of mismatch repair. Presence of MSI is associated with good prognosis in colorectal cancer patients, especially in stage II or III disease, and their presence, associated with other established prognostic factors, may obviate the need for adjuvant chemotherapy in patients with stage II disease.
Gene expression profiles such as the Oncotype Dx test, which measures expression of 21 genes in breast tumor tissue, enable calculation of a recurrence score that predicts the risk of distant disease at 10 years in a specific subset of breast cancer patients.
Prediction of treatment response A test that can identify whether or not a potential treatment is likely to be effective and of benefit to the patient. Measurement of estrogen receptors (ER) is mandatory for all newly diagnosed invasive breast cancers , to predict response to treatment with antiestrogen therapy (i.e., aromatase inhibitors and tamoxifen). The additional measurement of progesterone receptors (PR) can improve the accuracy of prediction. ,
Measurement of human epidermal growth factor receptor 2 ( HER2 ) is essential to identify patients with HER2 -positive breast cancer who are likely to benefit from treatment with anti- HER2 monoclonal antibody therapy (e.g., trastuzumab). ,
Monitoring response during and/or shortly after treatment A test that assesses whether the tumor is responding to treatment, enabling withdrawal and/or change of ineffective treatment. In patients receiving treatment for ovarian cancer, a reduction of at least 50% in CA125 concentrations from a pretreatment sample has been defined as a response, provided it is confirmed and maintained for at least 28 days and if a pretreatment CA125 concentration was greater than twice the upper limit of the reference interval and measured within 2 weeks of the start of chemotherapy.
Biological response to systemic chemotherapy, as assessed by serial measurement of serum CEA and CA19-9 in patients receiving chemotherapy for colorectal cancer with liver metastases, agrees with response as assessed by radiology in over 94 and 91% of patients, respectively, suggesting that this could decrease the need for imaging studies, such as computerized tomography (CT), which require exposure of patients to significant doses of radiation.
Monitoring disease post-treatment to detect progression A test that reliably indicates earlier than clinical symptoms whether disease is progressing in patients with no evidence of disease post-therapy and/or in patients with detectable disease. Whether this information is helpful crucially depends on whether alternative therapies are available. Inclusion of CEA measurements in intensive follow-up strategies for patients with nonmetastatic colorectal cancer following curative surgery has been shown to improve overall survival and increase the detection of asymptomatic recurrences and hence the frequency of curative surgery attempted at recurrence. Such follow-up is also associated with earlier detection of recurrence.
AFP , α-Fetoprotein; CEA , carcinoembryonic antigen; hCG , human chorionic gonadotropin.

In order to achieve optimal use of tumor markers, an evidence-based approach to clinical decision-making is essential. , As discussed later in this chapter, careful consideration must be given to whether an individual tumor marker can fulfill requirements for each use (see Table 33.3 ). In practice, most are appropriate only in selected clinical circumstances. Many guidelines on tumor marker use ( Table 33.4 ), including those of the American Society for Clinical Oncology (ASCO) and the European Society for Medical Oncology (ESMO), focus primarily on clinical management, with relatively little mention of tumor marker or other laboratory tests.

TABLE 33.4
Selected Expert Groups That Have Developed Guideline Recommendations Relevant to Tumor Marker Use
Abbreviation Name of Organization Website
ASCO American Society for Clinical Oncology http://jco.ascopubs.org/site/misc/specialarticles.xhtml (Accessed November 2020)
EGTM European Group on Tumor Markers
ESMO European Society for Medical Oncology http://www.esmo.org/Guidelines (Accessed November 2020)
NACB National Academy of Clinical Biochemistry https://www.aacc.org (Accessed November 2020)
NCCN National Comprehensive Cancer Network http://www.nccn.org (Accessed November 2020)
NICE National Institute for Health and Care Excellence https://www.nice.org.uk/guidance (Accessed November 2020)
SIGN Scottish Intercollegiate Guideline Network http://www.sign.ac.uk/ (Accessed November 2020)

Guidelines published by the National Comprehensive Cancer Network (NCCN) in the United States include patient pathways that clearly indicate which tumor markers should be measured and when. Complementing these clinically oriented guidelines, those published by the National Academy of Clinical Biochemistry (NACB) and the European Group on Tumor Markers (EGTM) focus on the use of tumor markers, considering in detail requirements for their appropriate use in all phases of both the patient pathway and laboratory provision, helpfully comparing recommendations made with those of other groups. Recommendations relating to tumor marker use for specific malignancies are summarized later in this chapter.

While there have been many reports on tumor markers in the last 50 years—the number of publications on “neoplastic antigens” increased from about 260 in 1973 in Index Medicus to more than 348,700 in 2020 in PubMed —the number of tumor markers with established roles in clinical practice has remained rather low. Only a few new serum and tissue markers have been introduced into routine practice in the last 25 years, , although there have been major developments in fecal testing for colorectal cancer. (See the section on “Colorectal Cancer.”) Assessments of BRAF mutations for melanoma and EGFR mutations for non–small cell lung cancer (NSCLC) are now routine in clinical practice. (See sections on Melanoma and Lung cancers).

Well-established and commonly requested serum tumor markers whose measurement is generally available in most large clinical laboratories are listed in Table 33.5 , along with some of their properties and the cancers with which they are primarily associated. Some less commonly requested serum tumor markers with an established clinical role are shown in Table 33.6 . Table 33.7 lists some of the tissue markers whose measurement enables informed decisions regarding selection of endocrine and immunotherapy and other targeted therapies in individual patients. Some of the many other tumor markers that have not found clinical application are tabulated elsewhere. , ,

TABLE 33.5
Properties and Applications of Commonly Requested Serum Tumor Markers
Tumor Marker Biochemical Properties Molecular Weight Main Clinical Applications
Alkaline phosphatase Phosphohydrolase Variable Raised activities associated with presence of liver and/or bone metastases
Alpha-fetoprotein (AFP) Glycoprotein, ~4% carbohydrate; considerable homology with albumin ~70 kDa Diagnosis and monitoring of primary hepatocellular carcinoma, hepatoblastoma, and germ cell tumors. Prognosis of germ cell tumors.
Cancer antigen 125 (CA125) Mucin identified by monoclonal antibodies OC125 and M11; developed from serous cystadenocarcinoma cell line ~200 kDa Monitoring ovarian carcinoma. Measurement required for determination of the “Risk of Malignancy index” (RMI) for ovarian carcinoma.
Carcinoembryonic antigen (CEA) Family of glycoproteins, 45% to 60% carbohydrate ~180 kDa Monitoring colorectal adenocarcinomas
ConfirmMDx A multiplex epigenetic assay Not applicable Reducing unnecessary repeat prostatic biopsies in men who have had at least one negative biopsy
Human chorionic gonadotropin (hCG) Glycoprotein hormone consisting of two noncovalently bound subunits (α and β). α-Subunit similar to LH, FSH, and TSH; β-subunit considerable homology with LH ~36 kDa Diagnosis, prognosis, and monitoring germ cell tumors and gestational trophoblastic neoplasia
Lactate dehydrogenase (LDH) Enzyme of the glycolytic pathway Variable Diagnosis, prognosis, and monitoring of germ cell tumors. Used to monitor a wide range of malignancies, including hematologic malignancies.
Paraproteins Monoclonal immunoglobulins Variable See Chapter 31 .
Prolactin Pituitary hormone ~22 kDa, but high molecular forms also exist See Chapter 55 .
Prostate-specific antigen (PSA) Glycoprotein; member of the kallikrein family with serine protease activity; circulates as free enzyme or complexed to α 1 -antichymotrypsin (measurable) or α 2 -macroglobulin (not detected by most immunoassays) ~30 kDa (free enzyme) Diagnosis, risk assessment, and monitoring of prostate carcinoma. Lower concentrations of free PSA relative to complexed PSA (i.e., free: total ratio) found in prostatic cancer as compared with benign prostatic hypertrophy.

TABLE 33.6
Properties and Applications of Less Commonly Requested Serum Tumor Markers
Tumor Marker Properties Molecular Weight Main Clinical Applications
Calcitonin 32-amino acid peptide ~3.5 kDa Monitoring medullary carcinoma of the thyroid
Cancer antigen 15.3 (CA15.3), BR 27.29 Mucin (MUC-1 glycoprotein peptide) i dentified by monoclonal antibodies >250 kDa Monitoring breast cancer
Cancer antigen 19.9 (CA19.9) Glycolipid carrying the Lewis a blood group determinant ~1000 kDa Monitoring pancreatic carcinoma following curative resection
Catecholamines Biogenic amines ~0.2 kDa See Chapter 53
Chromogranin A Member of the granin family of acidic secretory glycoproteins ~49 kDa Monitoring neuroendocrine tumors
CYFRA 21-1 Fragments of cytokeratin 19 ~30 kDa Monitoring lung carcinoma
Gut hormones Small peptide hormones, including vasoactive intestinal peptide (VIP), pancreatic polypeptide (PP), somatostatin, gastrin See Chapter 52
Human epididymis protein 4 (HE4) Product of the WFDC2 (HE4) gene that is overexpressed in patients with ovarian carcinoma ~25 kDa Monitoring ovarian cancer. Potentially an aid in diagnosis as part of the risk of malignancy algorithm (ROMA) (207). Under evaluation.
Inhibin A (α-β A ) Inhibin B (α-βB) Heterodimeric glycoproteins composed of an α- and a β-subunit. There are several forms of the β-subunit, as indicated 32 kDa Monitoring of ovarian granulosa cell tumors and testicular Sertoli and Leydig tumors
β 2 -microglobulin Component polypeptide chain of the HLA antigen complex ~11 kDa Providing prognostic information for patients with multiple myeloma
Neuron-specific enolase (NSE) Dimer of the enzyme enolase ~87 kDa Monitoring small cell lung carcinoma, neuroblastoma, and neuroendocrine tumors
Placental alkaline phosphatase (PLAP) Heat-stable isoenzyme of alkaline phosphatase ~86 kDa Monitoring of germ cell tumors (seminomas)
Pro-gastrin releasing peptide (proGRP) More stable precursor of gastrin releasing peptide ~16 kDa Monitoring small cell lung carcinoma
Prostate cancer gene 3 protein (PCA3) Protein product of PCA3 gene in urine of prostate cancer patients ~86 kDa Potentially an aid to diagnosis of prostate cancer, particularly in biopsy-negative men. Under evaluation.
Prostate marker algorithm Phi Not applicable Not applicable An algorithm combining serum measurements of total PSA, free PSA, and proPSA in serum
Prostate marker algorithm 4K Not applicable Not applicable An algorithm combining serum measurements of free and total PSA, human kallikrein 2, and intact PSA
Squamous cell carcinoma antigen (SCC) Serine protease inhibitor isolated from glycoprotein subfraction of tumor antigen TA-4 ~48 kDa Monitoring squamous cell carcinomas (e.g., cervix)
S100 proteins Family of proteins characterized by two calcium-binding sites that have helix-loop-helix conformations ~20 kDa Monitoring malignant melanoma
Thyroglobulin (Tg) Glycoprotein dimer of two identical subunits ~670 kDa Monitoring differentiated thyroid cancer
Tissue polypeptide antigen (TPA) Fragments of cytokeratins 8, 18, and 19 ~22 kDa Monitoring bladder and lung carcinoma

TABLE 33.7
Tissue Markers Whose Measurement Informs Selection of Endocrine, Immunotherapy, and Other Targeted Therapies in Individual Patients
Tumor Marker Biochemical Properties Molecular Weight Main Clinical Applications
Anaplastic lymphoma receptor tyrosine kinase (ALK) Enzyme encoded by the ALK gene ~176 kDa Predicting response to the thymidine kinase inhibitor (TKI) crizotinib in non–small cell lung cancer
BRA mutation Proto-oncogene that encodes the serine/threonine protein kinase B-Raf ~87 kDa Predicting response to inhibitors of BRAF-mutated protein, including vemurafenib and dabrafenib in melanoma
Epidermal growth factor receptor (EGFR) Transmembrane protein receptor involved in cell signaling pathways ~170 kDa
Estrogen receptor (ER) Nuclear transcription factor ~5 kDa Predicting response to endocrine therapy in breast cancer
Human epidermal growth factor receptor-2 ( HER2 or c-erb2) Transmembrane glycoprotein encoded from HER2 /neu oncogene ~185 kDa Predicting response to Trastuzumab (Herceptin) in breast cancer.
Predicting response to tyrosine kinase inhibitors (TKIs) in non–small cell lung cancer.
KRAS mutation Guanosine-nucleotide-(GTP)-binding protein ~23 kDa Predicting response TKIs in non–small cell lung cancer and reducing need for testing for HER2 and ALK alterations
Progesterone receptor Nuclear transcription factor A form ~4 kDa
B form: ~120 kDa
Predicting response to endocrine therapy in breast cancer

The broad principles guiding the validation of any new tumor marker and its subsequent introduction into routine clinical practice are essentially similar and are considered in detail in the following sections. Early in the evaluation of a promising new tumor marker, critical aspects include rigorous validation of analytical performance and clinical utility and demonstration of clinical value. While in the past tumor markers may have been prematurely introduced into clinical use without appropriate evaluation, objective assessment of the potential clinical value of a proposed new tumor marker, together with an estimate of the magnitude of its benefit, including effect on patient outcome, is now a prerequisite for introducing a new test into routine practice. Three key issues in tumor marker evaluation are clinical utility, magnitude of effect, and reliability. , Only when these have been established and regulatory requirements fulfilled should the tumor marker be adopted into routine clinical practice. It is then essential to ensure that the test is requested appropriately, that those requesting the test are aware of any preanalytical requirements, that these are met in routine practice, that analytical performance is reliable as confirmed by rigorous internal quality control (IQC) and proficiency testing (PT), and that postanalytical reporting of results to clinical users is both informed and informative.

Tumor marker validation

The steps required to take a new tumor marker into routine use—from the research laboratory to the specialist laboratory and then into routine clinical practice—have been described in detail. , A position statement from the EGTM provides detailed recommendations about the essential steps in the validation of a tumor marker. Criteria for the use of omics-based predictors in clinical trials have also been published by a panel convened by the US National Cancer Institute. , Assuming there is preliminary evidence that the marker has clinical potential, there are six requirements for validation prior to introduction into routine use. These are detailed in the following sections.

Protocols for sample collection and processing

Availability of clinical specimens (e.g., serum, plasma, tissue) that have been collected, processed, and stored following validated standard operating procedures (SOP) that take account of potentially confounding factors, including those described below, is essential. , The panel should include samples from patients with a variety of malignant and nonmalignant conditions. Those from cancer patients should ideally include samples from all stages of disease and especially from patients with early-stage disease. Specimens from biobanks may be appropriate provided their provenance is well documented.

Analytical/technical validation

Each specific assay that will be used in the clinic must be developed and validated, having regard to the requirements described below (see the section on “Analytical Requirements”). Metrological requirements were established at the Stockholm Conference in 1999 and have recently been updated. , These incorporate assessments in which components of biological variation of the marker are taken into account where feasible. Detailed guidance on analytical requirements for tumor markers is available for serum-based immunoassays and immunohistochemical tests. ,

Clinical validation

Defining the specific context in which the tumor marker is to be used is an essential first step that will identify the population to be used in the clinical validation step (i.e., the type of specimens required from carefully defined groups of subjects), the informative statistics, and the acceptability criteria. , Clinical validation should be reported in terms of diagnostic accuracy for tests intended for use in diagnosis, as described in Tables 33.8 and 33.9 . Pitfalls in clinical validation include study bias, overfitting of data, and multiplicities (statistical problems that may occur—for example, with disease subset analysis or the use of several different endpoints). The risk of these can be decreased by strict adherence to a written protocol and reporting of all planned steps and all work completed. The EGTM has proposed a four-phase model for tumor marker monitoring trials that is analogous to those used for the investigation of new drugs. Biomarker kinetics and correlation with tumor burden are assessed in phase I, while in phase II, the ability of the marker to identify, exclude, and/or predict a change in disease status is evaluated. In phase III the effectiveness of intervention based on trends in tumor marker concentration is assessed by measuring patient outcome in randomized trials, while phase IV involves clinical evaluation of long-term effects following incorporation of the tumor marker into routine clinical care. The first two phases are also applicable to tumor markers intended to be used solely for screening or diagnosis.

TABLE 33.8
Parameters Used to Describe and Validate the Clinical Performance of a Tumor Marker
Parameter Description Calculation
Clinical sensitivity How successfully the test identifies individuals who have the condition TP/(TP + FN)
Clinical specificity How successfully the test excludes individuals who do not have the condition TN/(TN + FP)
Prevalence How common the condition is in the study cohort (TP + FN)/(TP + FN + TN + FP)
Positive predictive value (PPV) or positive likelihood ratio How much more likely the test is to be positive in a patient with the condition than in a person without it TP/(TP + FP)
Negative predictive value (NPV) or negative likelihood ratio How much more likely the test is to be negative in a patient without the condition than in a person with it TN/(TN + FN)
Accuracy How likely it is that the test will correctly identify the presence or absence of a cancer (TP + TN)/(TP + TN + FP + FN)
Receiver operating characteristic (ROC) curve (see Fig. 33.3 ) Graphical representation of test performance over a range of test concentrations
Area under the (ROC) curve (AUC) Numerical means of comparing different tests
FN, False negative; FP, false positive; TP, true positive; TN, true negative.

TABLE 33.9
Example of Calculations for Data From 100,000 Women Post–Hydatidiform Mole Using Human Chorionic Gonadotropin to Detect Choriocarcinoma
Modified from Roulston JE, Leonard RCF. Serological tumour markers. Edinburgh: Churchill Livingstone, 1993. Used with permission.
Positives Negatives Totals
hCG positive 7920 (TP) 920 (FP) 8,840 (TP + FP)
hCG negative 80 (FN) 91,080 (TN) 91,160 (FN + TN)
Totals 8000 (TP + FN) 92,000 (TN + FP) 100,000 (All)
Sensitivity = TP/(TP + FN) = 99%
Specificity = TN/(TN + FP) = 99%
Prevalence = (TP + FN)/(TP + FN + TN + FP) = 8%
Positive predictive value = TP/(TP + FP) = 89.6%
Accuracy = (TP + TN)/(TP + TN +FP + FN) = 99%
FN, False negative; FP, false positive; TP, true positive; TN, true negative.

Demonstration of clinical value

Strong evidence that the test will benefit the patient (e.g., by increasing overall survival or leading to fewer invasive procedures or hospital visits) is now mandatory for introduction of new tests into most health care systems. Ideally this should be high-level evidence from a prospective or retrospective randomized clinical trial or systematic review of the literature.

Achieving regulatory approval

Requirements vary in different countries but in the United States involve either clearance approval by the US Food and Drug Administration (FDA) and evaluation in a Clinical Laboratory Improvement Amendments (CLIA)-certified laboratory or by a laboratory-developed test (LDT) pathway.

Postmarket evaluation

Analytical performance of the test in the routine clinical setting should be subject to continued evaluation, best achieved through implementation of rigorous IQC and PT procedures. Ongoing clinical audit to ensure that the marker retains its clinical value and that it is being used in the setting in which it was originally validated is also essential. When a newly introduced tumor marker replaces an earlier one, it is essential to ensure that the first test is discontinued.

“Ideal” tumor marker

In view of the above, it is helpful to consider requirements of an “ideal” tumor marker, which include the following:

  • 1.

    Detectable only in a given malignancy and absent in the healthy population or in nonmalignant conditions (i.e., with clinical specificity and clinical sensitivity approaching 100%).

  • 2.

    Present in a readily acquired biological matrix (e.g., serum, urine, tumor tissue).

  • 3.

    Present at concentrations proportional to tumor burden.

  • 4.

    Conveniently measured by a readily available, simple, reproducible, and inexpensive procedure.

  • 5.

    Beneficial to clinical care with a measurable effect on patient outcome.

No such tumor marker has yet been identified. However, measurement of hCG approaches ideal in the screening and monitoring of women who have had a molar pregnancy and are at risk of developing gestational trophoblastic neoplasia (GTN) (choriocarcinoma) (see the section on “Screening for Cancer”). Other tumor markers can also contribute usefully to patient management provided they are used intelligently and with regard to requirements of the clinical application (see Table 33.3 ) as outlined below.

Clinical applications of tumor markers

Screening for cancer

Screening for disease differs fundamentally from diagnosis or “case finding” because the aim of screening is to detect unsuspected disease in asymptomatic subjects (see Table 33.3 ). The World Health Organization (WHO) criteria for screening programs originally proposed by Wilson and Jungner in 1968 remain valid. Table 33.10 compares how well these criteria are met by a long-established national screening program for choriocarcinoma in high-risk women and by proposed screening programs for prostate cancer. High sensitivity and high specificity of the test for the target disease are essential. Also fundamental to the success of the choriocarcinoma screening program is the high prevalence of the condition (estimated as 3 to 10%) in the high-risk population being screened, yielding a positive predictive value of approximately 92%. The important effect of prevalence of disease on positive predictive value is further illustrated in Table 33.11 .

TABLE 33.10
WHO Criteria for Screening and How Well These Are Met by Established Screening Programs
WHO Requirements Example: Screening for Choriocarcinoma in a Selected Population (Already Est. in the UK ) Example: Screening for Prostate Cancer (Under Evaluation Nationally and Internationally)
Disease
Should be an important health problem Accounts for 0.02% of cancer deaths, in women of childbearing age Second leading cause of cancer deaths among men in some Western countries
Should be serious Fatal if not treated Mortality rate significant if not treated
Should have a recognizable early (premalignant) stage Previous partial or complete hydatidiform mole in previous pregnancy. (Screening restricted to women who have had such a pregnancy.) None identified. (PSA may be increased in benign prostatic hyperplasia, which is not precancerous.)
Should have a natural history that is well understood Steps in progression reasonably well documented Natural history reasonably understood, but what differentiates aggressive from indolent tumors unknown. Significant risk of overdetection.
Test or Examination
Should be accurate and reliable hCG assays have ~99% specificity and sensitivity for the disease. (hCG is not detectable in serum in healthy nonpregnant subjects.) In a population of 63-year-old men at a cutoff point of 4.0 μg/L, PSA had a sensitivity of 46% and a specificity of 91% for identifying prostate cancer that was clinically important within the next 10 years
Should be acceptable to the population being screened Screening test can be performed on urine specimens that can be conveniently collected at home by the patient and sent by mail to the screening center Blood sampling acceptable to most patients
Should be part of an ongoing surveillance system A national registration system can be established for patients at risk Recall systems likely to be complex to administer
Should be repeated at intervals if desirable (depending on the natural history of the disease) Lifetime follow-up recommended because disease can recur years later Benefit of repeat testing still being assessed
Relating to Treatment of Identified Disease
Early intervention should be of more benefit than treatment started at a later stage Prompt treatment with chemotherapy is highly desirable Benefit of treatment not proven yet except in men with localized disease who are candidates for radical prostatectomy. In these men the absolute reduction in the risk of death after 10 years is small, but the reduction in the risks of metastasis and local tumor progression are substantial. Watchful waiting in some patients is as appropriate as active intervention.
There should be an acceptable and effective treatment for patients with recognized disease Chemotherapy is highly effective Radical prostatectomy may be curative in disease confined to the prostate. See above comment also.
There should be appropriate facilities for diagnosis and treatment Three specialist centers undertake testing in the United Kingdom, with all patients referred for treatment to Charing Cross Hospital, London Significant requirements for additional diagnostic imaging, urologists, counselors, and so on
There should be an agreed policy about whom to screen and treat All women with a previous history of a molar pregnancy should be screened General agreement that men over 50 years old (or younger if family history) may benefit from screening. How to select those who will benefit from treatment not known.
The chance of physical or psychologic harm should be less than the chance of benefit Treatment is lifesaving Physical side effects of surgical and/or radiation treatment include incontinence and impotence in a significant number of patients. Psychologic anxiety associated with a positive test and the uncertainty about whether screening reduces the risk of death from prostate cancer.
Cost of Screening Program
Should be balanced against the benefits it provides Costs of the screening test and program are relatively low compared with the social cost avoided per life saved Accurate cost analysis not yet available but likely to be considerable; should include costs of patient education, advertising for screening, PSA test, biopsy, pathology, staging, definitive therapy, secondary therapy, monitoring, and treatment of complications.

TABLE 33.11
Effect of Prevalence of the Predictive Value of a Positive Test
Modified from Roulston JE, Leonard RCF. Serological tumour markers. Edinburgh: Churchill Livingstone, 1993. Used with permission.
POSITIVE PREDICTIVE VALUE
Disease Prevalence (%) Test Sensitivity and Specificity = 95% Test Sensitivity and Specificity = 99%
0.02 0 2.0
0.1 1.9 9.0
1.0 16.1 50.0
2.0 27.9 66.9
5.0 50.0 83.9
8.0 62.2 89.6
50.0 95.0 99.0

The relevant test parameters described in Table 33.8 are depicted schematically in Fig. 33.2 . Diagnostic accuracy is established by studying test performance at different decision (cutoff) concentrations in appropriately selected diseased and disease-free populations. The relationship between these parameters is most conveniently depicted graphically by a receiver operating characteristic (ROC) plot ( Fig. 33.3 ). Changing the decision concentration selected alters both sensitivity and specificity but cannot improve both concurrently. These parameters define the characteristics of the test and, together with the prevalence of the disease in the population studied, yield the positive predictive values ( Box 33.1 ). The low prevalence of the majority of cancers, even in age groups most at risk, together with the low sensitivity and specificity of most tumor markers, generally precludes their use in screening. However, successful population screening for early colorectal cancer using a fecal immunochemical test for hemoglobin (FIT) is routine in some countries. The efficacy of screening patients at high risk of HCC and ovarian cancer is being actively investigated, and there is continuing debate about whether to screen for prostate cancer. The possibility of implementing genetic screening for some conditions is also being considered. These issues are considered in more detail in the organ-specific sections in this chapter.

FIGURE 33.2, The effect of tumor marker cutoff concentration on sensitivity and specificity (proportion of true negatives) and sensitivity (proportion of true positives) in diseased and disease-free populations.

BOX 33.1
Modified from Roulston JE, Leonard RCF. Serological tumour markers. Edinburgh: Churchill Livingstone, 1993. Used with permission.
Example Calculation Showing the Effect of Prevalence on Positive Predictive Value for Choriocarcinoma Detection With a Previous Molar Pregnancy

Assuming a disease prevalence of 8%

  • Of 1000 a women screened:

  • Number with disease = 80 (0.08 × 1000), so with a specificity of 99%, 79.2 (80 × 0.99) cases correctly identified (true positives)

  • Number without disease = 920 (0.92 × 1000), so with a sensitivity of 99%, 9.2 [920 × (1.0−0.99)] negative cases are incorrectly identified as having the disease (false positives)

  • The positive predictive value (probability) of obtaining a positive result in a patient with choriocarcinoma is therefore [79.2/(79.2 + 9.2)] × 100 = 89.6 (An excellent test)

Assuming a disease prevalence of 1%

  • Of 1000 a women screened:

  • Number with disease = 10 (0.01 × 1000), so with a specificity of 99%, 9.9 (10 × 0.99) cases correctly identified (true positives)

  • Number without disease = 990 (0.99 × 1000), so with a sensitivity of 99%, 9.9 [990 × (1.0−0.99)] negative cases are incorrectly identified as having the disease (false positives)

  • The positive predictive value (probability) of obtaining a positive result in a patient with choriocarcinoma is therefore [9.9/(9.9 + 9.9)] × 100 = 50.0 (Still a good test but not as good as the case when the prevalence is higher)

a In 1000 women, assuming test specificity and sensitivity are both 99%.

Diagnosis of cancer

The same limitations of lack of sensitivity and specificity that apply to the use of tumor markers for screening also apply to diagnosis and case finding (see Table 33.3 ). Tumor markers are not helpful for diagnosis in patients with nonspecific symptoms and cannot replace a biopsy for establishing the primary diagnosis of cancer. However, in carefully selected undiagnosed patients who are at high risk of malignancy, raised concentrations of tumor markers may be informative and facilitate diagnosis (e.g., when the patient is unable or unwilling to undergo more invasive testing such as colonoscopy). In general, the higher the serum tumor marker value, the greater the likelihood of malignancy, but conversely it is essential to remember that results within the reference interval never necessarily exclude malignancy. The NACB recommends that requests for panels of tumor marker measurements be actively discouraged, together with inappropriate requests for prostate-specific antigen (PSA) in women or CA125 in men, because these are unlikely to lead to improved patient outcomes.

FIGURE 33.3, Receiver operating characteristic (ROC) plot from which assay performance is assessed by plotting sensitivity as a function of (1—specificity). A highly discriminatory test will give a curve of steep slope from the origin, while a random test will give a straight line as indicated.

Assessing prognosis

Prognostic markers predict the likely outcome of disease with respect to risk of relapse or disease progression (see Table 33.3 ). They are generally most useful at the time of initial diagnosis, when a marker of good prognosis is suggestive of prolonged survival and/or the possibility of cure. A marker of poor prognosis indicates an increased probability of early recurrence. Prognostic markers can help identify patients with aggressive tumors that require further treatment (e.g., with systemic adjuvant chemotherapy following surgery) while minimizing the risk of overtreatment of patients with indolent disease that is unlikely to progress. It is important to note that prognostic markers provide a probability estimate of outcome for a heterogeneous population of patients and that no prognostic marker can accurately predict outcome for an individual patient.

Although many tumor markers have been reported to have prognostic significance, with more than 22,000 references relating to cancer prognostic markers in PubMed, fewer than 10 are regularly used in clinical practice. Possible reasons for this include inadequate study design, use of inappropriate statistical tests and overoptimistic reporting, failure of a marker to provide independent prognostic information adding to that from established prognostic factors (e.g., tumor size, grade, and lymph node involvement), and inadequate validation. Most prognostic markers are measured in tissue, but as indicated in Table 33.3 , measurement of AFP, hCG, and LDH following primary surgery in patients with nonseminomatous germ cell tumors (GCT) is essential. These markers provide strong independent prognostic information complementing that of conventional prognostic factors and have been well validated. Additionally, although prognostic markers may be of limited use unless there is a therapy that can change the course of the disease, some patients and caregivers may find prognostic information helpful.

Prediction of treatment response

The heterogeneous nature of cancer means that even cancers of the same histologic type may vary widely in response to a particular treatment, and for most cancers only a subset of patients will respond (see Table 33.3 ). Predictive markers that can identify those who will respond are therefore critical in treatment planning so that treatment can be offered to patients who will benefit and so alternative treatment can be offered to those who will not, also sparing them unnecessary side effects. With the development and use of new and costly molecularly targeted treatments, predictive markers (most of which are measured in tissue rather than serum) are increasingly both a clinical and an economic necessity. Several important predictive markers for breast cancer are already well established in clinical use. Further promising predictive markers are now available for breast cancer, colorectal cancer, glioma, melanoma, and NSCLC, while others are being investigated. (See the sections on Use of markers in specific malignancies.)

Monitoring response during and/or shortly after treatment

While the use of predictive tumor markers is increasingly important, the main application of most serum tumor markers is in monitoring the course of disease. For most diagnosed cancer patients, it is helpful to measure the relevant tumor marker pretreatment in order to provide a baseline for subsequent interpretation, and for some cancers, measurements made during and immediately after treatment are also desirable. In general, a decrease in marker concentrations to within expected population (i.e., “normal”) limits following treatment is a favorable sign. In some cases subnormal or absent post-treatment values should be achieved (e.g., for PSA postprostatectomy or thyroglobulin following thyroid ablation). Measurement of AFP and hCG is mandatory for GCTs prior to surgical excision because the pretreatment concentration is required to calculate the half-life of tumor marker decline following chemotherapy. Similarly, in ovarian cancer, measurement of CA125 and the extent of its decline during primary treatment may provide helpful prognostic information. (See the sections on specific cancers.)

Monitoring disease post-treatment to detect progression or recurrence

Serial monitoring of tumor marker concentrations post-treatment provides early indication of disease recurrence, often months before there are clinical signs and symptoms. Whether early detection is of benefit depends crucially on whether a rising tumor marker concentration will prompt clinical action (e.g., early ultrasound, CT or magnetic resonance imaging [MRI] scanning), whether an alternative treatment is available for the individual patient, and/or whether that treatment can be implemented without scan evidence of progression if none is available. If none of these apply and if the patient is not enrolled in a clinical trial, the patient should be made aware of the potential implications of having the test done, and whether tumor marker monitoring is likely to be of benefit should be seriously considered and discussed. ,

Quality requirements for tumor marker use in the clinic

While tumor marker results provide objective information that can facilitate clinical management, their appropriate selection and measurement are essential. The clinical laboratory should provide readily available information to those requesting tumor markers to encourage selection of the correct test or tests (and, more important, to discourage inappropriate requesting) and to ensure that specimen timing is appropriate and other preanalytical requirements are met. Ensuring that results are analytically correct and that accurate and informative reports are returned to the requesting clinician are also laboratory responsibilities. , Laboratory-oriented guidelines for tumor markers that outline quality requirements for provision of a high-quality tumor marker service have been developed by the NACB and the EGTM and form the basis of the following discussion.

Pre-preanalytical requirements

Although numerous serum tumor markers can be measured (see Tables 33.5 and 33.6 ), only those listed in Table 33.5 are commonly requested from the clinical laboratory. Table 33.12 summarizes the current recommendations of the NACB for their appropriate clinical use, , while typical clinical presentations that might prompt a request are described in Table 33.13 .

TABLE 33.12
Most Frequently Requested Serum Tumor Markers and National Academy of Clinical Biochemistry Recommendations for Their Appropriate Clinical Use
Modified from Sturgeon CM, Lai LC, Duffy MJ. Serum tumour markers: how to order and interpret them. BMJ 2009;339:b3527. Used with permission. Further information is available from the Trophoblastic Tumour Screening and Treatment Centre, Department of Medical Oncology, Charing Cross Hospital, London W6 8RF, UK (Tel: +44 (0) 20 8846 1409) or at http://www.hmole-chorio.org.uk/ . (Accessed November 2020).From Sturgeon CM, Duffy MJ, Stenman UH, et al. National Academy of Clinical Biochemistry laboratory medicine practice guidelines for use of tumor markers in testicular, prostate, colorectal, breast, and ovarian cancers. Clin Chem 2008;54:e11–79. Sturgeon CM, Duffy MJ, Hofmann BR, et al. National Academy of Clinical Biochemistry Laboratory Medicine Practice Guidelines for use of tumor markers in liver, bladder, cervical, and gastric cancers. Clin Chem 2010;56:e1–48.
CURRENTLY RECOMMENDED CLINICAL APPLICATIONS
Relevant Cancer Screening or Early Detection Diagnosis or Case Finding Prognosis (With Other Factors) Detecting Recurrence Monitoring Therapy
Alpha-fetoprotein (AFP) Germ cell/testicular tumor
Hepatocellular carcinoma a b c
Calcitonin Medullary thyroid carcinoma
Cancer antigen 125 (CA125) Ovarian cancer d e f
Cancer antigen 15-3 (CA15-3) Breast cancer g h
Cancer antigen 19-9 (CA 19-9) Pancreatic cancer i j
Carcinoembryonic antigen (CEA) Colorectal cancer c c
Human chorionic gonadotropin (hCG) Germ cell and testicular cancers; gestational trophoblastic neoplasia k
Paraproteins (M protein/Bence Jones protein) (also measured in urine) Multiple myeloma
Prostate-specific antigen (PSA) Prostate cancer
Thyroglobulin Thyroid cancer (follicular or papillary)

a Only for subjects in high-risk groups (e.g., with chronic HBV, HCV, or cirrhosis) and only in conjunction with ultrasound (impact on mortality unclear).

b In conjunction with liver imaging, AFP levels greater than 200 μg/L are regarded as virtually diagnostic of HCC in patients with hypervascular lesions.

c Especially for disease that cannot be evaluated by other modalities.

d Only for women at high risk of ovarian cancer and only in conjunction with transvaginal ultrasound.

e Only for differential diagnosis of pelvic masses, especially in postmenopausal women.

f Preliminary results of a randomized trial show no survival benefit from early treatment based on a raised serum CA125 level alone, so this recommendation may be modified to exclude asymptomatic patients.

g Postsurgery when it may provide lead time for early detection of metastasis but clinical value unclear.

h Especially in patients with nonevaluable disease (for which CEA is also recommended) in carefully selected patients.

i In patients in whom pancreatic disease is strongly suspected, CA19-9 may complement other diagnostic procedures.

j Especially after chemotherapy and in combination with imaging.

k Use of hCG in screening for gestational trophoblastic neoplasia, a rare malignancy that usually develops after a molar pregnancy, provides an excellent example of “best practice” in screening.

TABLE 33.13
Clinical Presentations That Might Prompt Requests for the Most Frequently Used Tumor Markers
Modified from Sturgeon CM, Lai LC, Duffy MJ. Serum tumour markers: how to order and interpret them. BMJ 2009;339:b3527. Used with permission.
Tumor Marker a Relevant Cancer Typical Clinical Presentation Other Cancers in Which the Marker May Be Raised b
Alpha-fetoprotein (AFP) Germ cell/testicular tumor Diffuse testicular swelling; hardness Gastric, colorectal, biliary, pancreatic, lung
Hepatocellular carcinoma Ascites, encephalopathy, jaundice; upper abdominal pain; weight loss; early satiety in high-risk subjects (i.e., hepatitis B or C–related cirrhosis) As above
Cancer antigen 125 (CA125) Ovarian cancer Pelvic mass; persistent, continuous, or worsening unexplained abdominal or urinary symptoms; bloating Breast, endometrial, cervix, peritoneal, uterus, lung, pancreas, hepatocellular, non-Hodgkin lymphoma
Cancer antigen 19-9 (CA19-9) Pancreatic cancer Progressive obstructive jaundice with profound weight loss and/or pain in the abdomen or midback Colorectal, gastric, hepatocellular, esophageal, ovary
Carcinoembryonic antigen (CEA) Colorectal cancer Intermittent abdominal pain, nausea, vomiting, or bleeding; palpable abdominal mass Breast, gastric, lung, mesothelioma, esophageal, pancreatic
Human chorionic gonadotropin (hCG) Germ cell/testicular tumor Diffuse testicular swelling, hardness, and pain Lung cancer
Gestational trophoblastic neoplasia Symptoms leading to x-ray showing cannonball secondaries; previous history of hydatidiform mole (see also Table 33.10 ) Lung cancer
Paraproteins (M protein/Bence Jones protein) (also measured in urine) Multiple myeloma Combination of symptoms including some/all of the following: anemia; back pain; weakness or fatigue; osteopenia; osteolytic lesions; raised ESR or globulins; spontaneous fractures; recurrent infections
Prostate-specific antigen (PSA) Prostate cancer Frequency, urgency, nocturia, dysuria; acute retention; back pain, weight loss, anemia None

a Tumor markers are often not helpful in diagnosis.

b This list is not comprehensive. The marker may be raised in other cancers.

Requests for tumor markers received in the clinical laboratory fall into three main categories: those for diagnosed cancer patients who have already been referred to specialist centers and are being or have been treated, those for patients who have been referred to a hospital for investigation of suspected malignancy and further investigation, and those requested by a family doctor or other primary care health professionals for patients who have presented with symptoms that could raise the suspicion of malignancy. Requests in the first category are likely to be made to monitor response to treatment (with a baseline measurement prior to therapy always desirable) or to detect recurrence and are most likely to be appropriate.

Nonspecialist users, whether in primary or secondary care, should always consider carefully whether a tumor marker result is likely to be helpful before the request is made. Requestors should be aware of the lack of sensitivity of most tumor markers, particularly for early-stage disease, their lack of specificity for a particular cancer, and the numerous nonmalignant conditions in which they may be increased (see Table 33.13 ). Nonspecialist users should also be aware that increased tumor marker concentrations do not necessarily indicate malignancy. Attempting to identify the reason for an increased tumor marker that should not have been requested and that is not associated with malignancy can be an expensive and time-consuming process, as well as psychologically stressful for the patient. Conversely, nonspecialist users should be reminded that whatever the malignancy or tumor marker, a result within the reference interval never necessarily excludes malignancy or progressive disease. Clinical biochemists should also think carefully before requesting additional tests that might lead to a diagnosis of malignancy and should seek the agreement of the clinician caring for the patient before doing so.

Unfocused requests such as “tumor marker screen” or “malignancy?” from emergency departments and other receiving units should be actively discouraged and met with offers of educational support. Reviewing requests prior to analysis is no longer feasible in most laboratories, but it can be helpful to provide readily available advice about appropriate test selection at the time of the request, together with reminders of their low sensitivity and specificity. This should be reasonably readily implemented when electronic requesting is available. Through the Pathology Harmony initiative in the United Kingdom, general advice that can be readily disseminated to nonspecialist users has been prepared in the form of a readily available and downloadable tumor marker bookmark. ,

Preanalytical requirements

Specimen timing

Timing of specimens within the day for tumor marker measurement is not usually critical because there is little evidence of diurnal variation for most markers. A pretreatment specimen is always helpful when interpreting subsequent results. In some cases, specific procedures may cause transient release of some tumor markers. Specimens should therefore be collected in advance of some procedures (e.g., CEA before colonoscopy or PSA before prostatic biopsy or digital rectal examination [DRE]) or after a suitable time interval after the procedure (e.g., CA125 following abdominal surgery). Awareness of benign conditions that may cause transient increases in tumor marker concentrations ( Table 33.14 ) and avoidance of inappropriate timing, if possible (e.g., measurement of CA125 in a woman who is menstruating, PSA in a man with an active urinary tract infection, or CA19-9 in a subject with cholestasis), reduce the risk of misinterpreting spuriously raised results that may cause undue distress to the patient and also decrease confidence in laboratory testing.

TABLE 33.14
Nonmalignant Conditions That Can Increase Serum Tumor Marker Concentration, Which May Lead to Incorrect Interpretation
Clinical Condition Tumor Markers a
Acute cholangitis CA19-9
Acute hepatitis CA125, CA15-3
Acute and/or chronic pancreatitis CA125, CA19-9
Acute urinary retention CA125, PSA
Arthritis/osteoarthritis/rheumatoid arthritis CA125
Benign prostatic hyperplasia (BPH) PSA
Cholestasis CA19-9
Chronic liver diseases (e.g., cirrhosis, chronic active hepatitis) CEA, CA125, CA15-3, CA19-9
Chronic renal failure CA125, CA15-3, CEA, hCG
Colitis CA125, CA15-3, CEA
Congestive heart failure CA125
Cystic fibrosis CA125
Dermatologic conditions CA15-3
Diabetes CA125, CA19-9
Diverticulitis CA125, CEA
Endometriosis CA125
Heart failure CA125
Irritable bowel syndrome CA125, CA19-9, CEA
Jaundice CEA, CA19-9
Leiomyoma CA125
Liver regeneration AFP
Menopause hCG
Menstruation CA125
Mesothelioma CEA
Nonmalignant ascites CA125
Ovarian hyperstimulation CA125
Pancreatitis CA125, CA19-9
Pericarditis CA125
Peritoneal inflammation CA125
Pregnancy AFP, CA125, hCG
Prostatitis PSA
Recurrent ischemic strokes in patients with metastatic cancer CA125
Respiratory diseases (e.g., pleural inflammation, pneumonia) CA125, CEA
Sarcoidosis CA125
Systemic lupus erythematosus CA125
Urinary tract infection PSA
AFP , α-Fetoprotein; CEA , carcinoembryonic antigen; hCG , human chorionic gonadotropin; PSA , prostate-specific antigen.

a This list is not comprehensive; these markers may be raised in other nonmalignant conditions.

Specimen handling

Serum or plasma is usually (but not always) equally appropriate for tumor marker measurements, although gel tubes may not be suitable for some assays. Requirements should always be checked in the product information supplied with the reagents and/or from other sources. The common tumor markers are generally stable, but serum or plasma should be separated from the clot and stored at 4 °C (short-term) or below −30 °C as soon as possible, following relevant guidelines where available. For longer-term storage, specimens should be stored at −70 °C. Heat treatment (e.g., to deplete serum complement components or to inactivate human immunodeficiency virus [HIV]) should be avoided, particularly for hCG (which may dissociate at increased temperature to form its free α- and β-subunits) and PSA. Potential influence of transit time on analyte results should be considered when samples are exposed to increased temperatures. Standardized conditions of specimen collection and fixation are crucial for immunohistochemical analyses.

Analytical requirements

Assay validation

Prior to their introduction into routine clinical practice, both immunoassays and immunohistochemical methods must be validated as described above by defined and well-characterized protocols that meet regulatory guidelines (e.g., FDA approval in the United States and CE marking in Europe). Individual laboratories should verify analytical performance prior to introduction into routine use. Internationally recognized guidelines for the performance of immunohistochemical tests should be adopted where these are available. If appropriate high-quality reference materials are not available, it is essential that methods for immunohistochemistry are described in detail.

Internal quality control

As for any laboratory tests, robust procedures for IQC should be established. Within-run variability less than 5% and between-run variability less than 10% should be readily achievable for automated tumor marker methods. Some newer techniques may perform significantly better than this, although manual and/or research assays may be less precise. Appropriate action should be taken immediately if an assay run fails to meet objective criteria for assay acceptance so no potentially erroneous results are reported. Criteria for acceptance should be predefined and preferably based on logical criteria such as those of Westgard. The number of IQC specimens included per run should allow identification of an unacceptable run with a given probability appropriate to the clinical application. Given the long-term monitoring involved in cancer care, assay stability should be ensured over prolonged periods. Laboratories should have in place procedures and acceptance criteria for assessment of lot-to-lot variation which may adversely affect clinical outcomes.

Quality control (QC) material not provided by the method manufacturer is preferable because 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. Negative and low positive controls should be included for all tumor markers and should include concentrations close to important decision points (e.g., 0.1 and 3 or 4 μg/L for PSA; 4 to 7 μg/L for AFP; 5 U/L for hCG). The broad concentration range should be covered, and ideally a high concentration control should occasionally be included to check the accuracy of dilution, whether manual or onboard.

Proficiency testing/external quality assessment

PT specimens should ideally be prepared from authentic patient sera, which for tumor markers may require dilution of high-concentration patient sera into a normal serum base pool. , As for IQC, specimens prepared by spiking purified analyte into serum base pools is likely to provide an overly optimistic impression of between-method performance. PT specimens should be commutable with patient specimens to ensure valid between-method comparisons. , Concentrations should assess performance over the working range and should include assessment of linearity on dilution, baseline security, and stability of results over time.

It is the responsibility of the PT provider to ensure that specimens are stable in transit. The target values (usually consensus means for heterogeneous analytes such as the tumor markers) should be accurate and stable as demonstrated by assessment of their accuracy (e.g., recovery of known amounts of added analyte), stability (i.e., reproducibility on repeat distribution of the same pool), and linearity on dilution (i.e., by issuing different dilutions of the same patient specimen). Because tumor markers are often monitored over long periods, regular assessment of reproducibility and stability of results over time is highly desirable. Reproducibility of results at low concentration is particularly important for AFP and hCG in GCTs and PSA in patients following prostatectomy because treatment may be instituted solely on the basis of a small increase in tumor marker concentration.

Occasional specimens should ideally be distributed to assess whether interference is observed in different methods (e.g., from biotin, heterophilic and other antibodies, high-dose hooking [see section below on “Clinically Relevant Interferences”]). Evaluation of interpretation and technical results is required for PT of immunohistochemical tests and can also be provided for quantitative tests through interpretative exercises and surveys of practice. These can make a powerful contribution to national audit by highlighting differences in reference intervals, reporting practice, and interpretation of clinical results, particularly when the ethos of the PT scheme is educational rather than regulatory.

Standardization

Major international efforts continue to be directed toward encouraging manufacturers to calibrate their methods accurately in terms of the established relevant International Standard (IS) or Reference Reagents ( Table 33.15 ). While availability of such standards does not guarantee improvement in between-method agreement, a reference material (provided its commutability can be demonstrated) provides a benchmark against which the accuracy of calibration can be assessed. Following adoption of the first IS for PSA by most providers of PSA assays, the mean between-laboratory CVs observed in the UK National External Quality Assessment Service (UK NEQAS) PT scheme for PSA decreased by approximately twofold, from more than 20% in 1995 to approximately 9.5% in 2005. Encouraging use of equimolar methods for PSA (i.e., PSA methods that recognize free and complexed PSA equally well) contributed to this improvement.

TABLE 33.15
WHO International Standards and Reference Reagents for Major Tumor Markers
Tumor Marker Code Year Established Description Reference
AFP IS 72/225 1972 Crude cord serum (50%)
CA125 No IS established
CA15-3 No IS established
CA19-9 No IS established
CA72-4 No IS established
CEA IRP 73/601 1973 CEA purified from liver metastases to primary colorectal cancer
hCG IS 07/364 2009 hCG, highly purified from human urine
hCGα IRP 75/569 1975 α-Subunit of hCG from human urine
hCGβ IRP 75/551 1975 β-Subunit of hCG from human urine
hCG IRR 99/688 2001 hCG, free from nicked forms and free subunits, highly purified from human urine
hCGn IRR 99/642 2001 Nicked hCG, partially degraded, missing peptide bonds in the hCGβ-40-50 region, highly purified from human urine
hCGα IRR 99/720 2001 α-Subunit of hCG, dissociated from hCG
hCGβ IRR 99/650 2001 Highly purified dissociated urinary hCGβ, free from intact dimeric hCG, hCGα, and hCGβn
hCGβn IRR 99/692 2001 Partially degraded nicked hCGβ, missing peptide bonds in the hCGβ-40-50 region
hCGβcf IRR 99/708 2001 Residues hCGβn-6-40, joined by disulfide bonds to hCGβn-55-92
PSA IRR 96/670 2000 90 : 10 ratio of bound : free PSA
fPSA IRR 96/668 2000 Purified free PSA
International Standards and Reference Reagents are available from the National Institute for Biological Standards and Control, Potters Bar, Herts, UK. Available at http://www.nibsc.org . (Accessed November 2020).
fPSA, Free prostate-specific antigen; hCG, human chorionic gonadotropin; hCG α , hCG β-subunit; hCG β , hCG β-subunit; hCGn, nicked hCG; hCG β n, nicked hCG β-subunit; hCG β cf, hCG β-core fragment; IRP, International Reference Preparation; IRR, International Reference Reagent; IS, International Standard; PSA, prostate-specific antigen.

Improved understanding of what is being measured in an assay for a heterogeneous tumor marker is critical if improved comparability is to be achieved, as illustrated by the number of hCG-related molecules in Table 33.15 . Some further progress has been made by organizing collaborative workshops to identify the more clinically appropriate antibody specificities, , and discussions are in progress about how to improve between-method comparability for the complex CA tumor markers for which no ISs have been established (see Table 33.15 ). It is helpful if manufacturers provide clear information about the specificity of the antibodies used in their methods and data on cross-reactivity that is readily comparable with that of other methods so users are aware of the differences.

Despite these efforts to improve comparability, the molecular heterogeneity of most tumor markers means that results obtained using different methods are not interchangeable, and considerable care is required in the interpretation of serial results obtained in more than one method.

Clinically relevant interferences

Tumor marker measurements are subject to the same interferences as all immunoassays (see Chapter 26 ), but clinical biochemists need to be aware of several that are of particular relevance.

High-dose “hooking.”

Because tumor marker concentrations can range over several orders of magnitude, protocols enabling identification of high-dose “hooking” are essential to minimize the risk of reporting erroneously low results, particularly in patients for whom markers are being measured for the first time. An example is shown in Fig. 33.4 . The risk of “hooking” can be reduced by using solid-phase antibodies of higher binding capacity, by using sequential assays that include a wash step, and by assaying specimens at two dilutions. Methods vary in their vulnerability to this interference.

FIGURE 33.4, High-dose hooking in tumor marker immunoassays. The extremely high tumor marker concentration blocks antibody binding sites in the immunoassay such that the apparent result “hooks” back onto the calibration curve and an erroneously low result may be reported.

Specimen carryover.

Specimen carryover is possible whenever high-concentration specimens are assayed, so it is desirable to check periodically for this possibility. Tumor markers such as hCG can range over five orders of magnitude, so carryover of 1/10,000 can still lead to a false-positive result in the following sample.

Interference from heterophilic or human antimouse antibodies.

Some patient sera contain anti-immunoglobulin antibodies (most often IgG) that may react with some antibodies used as reagents in immunoassays. High concentrations of human antimouse antibodies (HAMAs) may also be present in serum from cancer patients who have received treatment with mouse monoclonal antibodies for imaging or therapeutic purposes. If either type is present, results may be falsely high or low. Identifying the presence of interfering antibodies requires a high degree of clinical suspicion that a tumor marker result is not correct, which may be aided by having relevant clinical details available. Once suspected, possible interference can be investigated by assaying the specimen at several dilutions, by reassaying after treatment with a commercially available blocking agent, by adding further nonimmune mouse serum to the reaction mixture and reassaying, and/or by reassaying the specimen using a different method provided by a different manufacturer (using different antibodies) and preferably using a different methodology (e.g., RIA). Caution should be applied in interpretation. For example, linear dilution does not always exclude the presence of HAMA, while nonlinear dilutions are likely to indicate a possible interference.

Postanalytical requirements

Brief clinical information about the source of the suspected or diagnosed malignancy and the treatment stage (e.g., preoperative, postoperative, prechemotherapy) is highly desirable. This information should be recorded both in the laboratory computer and on the laboratory report, which should include cumulative and, if possible, graphical reporting of serial results because trends in tumor marker results are generally more informative than single results. Such reporting facilitates interpretation of results and can also help to identify occasional errors in requesting or in the laboratory (e.g., incorrect sample identification or missampling on an analyzer). Cumulative or graphic display of results also highlights unexpected results (e.g., sudden changes that are out-of-accord with the clinical picture) that require confirmation and further investigation. Brief comments relating to interpretation of the analytical results (e.g., whether or not an increase is likely to be clinically significant) and helpful advice about the frequency of monitoring and the need for confirmatory specimens are also desirable.

Urgent results that may be required for immediate patient management should be identified by the reporting biochemist so as to ensure that they reach the relevant clinician promptly (e.g., by telephone if appropriate). These include tumor marker results that can be used to diagnose advanced disease in critically ill but treatable patients (e.g., AFP in hepatoblastoma, hCG in choriocarcinoma, AFP and hCG in nonseminomatous GCTs, and PSA in men with advanced prostate cancer that may respond to endocrine therapy). The consequences of failure to ensure communication of such results directly to the clinician responsible for the care of the patient can be severe. ,

In view of the method-related differences discussed above, the method used should be stated on the clinical report so any changes in method are readily identifiable. If there has been a method change, it is highly desirable that the laboratory indicates whether this is likely to influence 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 prior to the change. Managing the change may necessitate analyzing the previous specimen by the new method or by requesting a further specimen to reestablish the baseline and/or confirm the trend in marker concentrations. If the results are likely to be significantly affected by the change in method, as should be clear from the initial validation, it may be desirable to run old and new methods in parallel for a defined changeover period, an approach that also helps clinicians become accustomed to the new values.

Reference intervals should be derived using an appropriate healthy population and should be specific to the method used (see Chapter 9 ). They are usually most relevant for pretreatment of cancer patients. Subsequently, the patient’s individual baseline results provide the most important reference point for most marker results, and application of reference intervals derived in healthy populations can be misleading. For example, at least 6 weeks postprostatectomy, a confirmed PSA concentration of 2.0 μg/L suggests persistent or progressive disease, although this concentration is well within reference intervals for healthy individuals. Provided baseline marker concentrations are well established in post-treatment of diagnosed patients, sustained increases even within the reference interval or other decision limits may be significant and should be treated as possible relapse, provided the measurement procedure is the same.

Reporting critical increases in tumor marker concentrations, taking into account the analytical performance of the test, biological variation (where possible), and the individual reference intervals, helps to contribute to an earlier diagnosis of relapse. The percentage increase or decrease that constitutes a significant change should be defined and should take account of analytical and biological variation, , as well as the expected rate of change in benign and malignant conditions and the time between samples. For tumor markers, differences in the magnitudes of their biological variation contribute significantly to these percentages. , A confirmed increase or decrease of ±25% is frequently considered to be of clinical significance, but more work is required in this area.

Laboratories should be able to provide calculated tumor marker half-lives or doubling times for markers for which these are relevant (e.g., AFP and hCG). Half-lives are defined as the time to 50% reduction of circulating tumor marker concentration following complete removal of tumor tissue. Their calculation may be irrelevant if a 50% reduction does not represent a significant change. (See the section on “Germ Cell Tumors” for calculation details.)

Proactive provision of a high-quality tumor marker service helps to encourage good communication between laboratory and clinical staff and is likely to encourage appropriate use of tumor marker tests and early identification of any results that are not in accord with the clinical picture and require investigation. An example of a laboratory report that fulfills many of these requirements is shown in Fig. 33.5 .

FIGURE 33.5, Desirable clinical laboratory report for tumor markers that fulfills current reporting recommendations.

POINTS TO REMEMBER

Cancer and tumor markers

  • Cancer is a heterogeneous disease that is a leading cause of death, although death rates for individual cancers vary markedly, and considerable advances in treatment have been made over recent decades.

  • Tumor markers are surrogate indicators that can help make or confirm a cancer diagnosis, monitor treatment effectiveness and the course of disease, estimate prognosis, and/or predict whether a specific therapy is likely to be effective.

  • Optimal use of tumor markers requires an evidence-based approach to clinical decision making and knowledge of the limitations of these tests particularly in relation to clinical sensitivity and specificity.

  • Tumor marker results are rarely diagnostic and cannot replace biopsy for the primary diagnosis of cancer.

  • Tumor marker measurements are not recommended for patients with vague symptoms when the population likelihood of cancer is low.

  • A raised tumor marker result never necessarily indicates malignancy, and, conversely, a result within the reference interval never necessarily excludes malignancy.

  • In diagnosed patients a pretreatment result is essential and provides the baseline against which subsequent results can be assessed.

  • Tumor marker results should always be confirmed on a repeat specimen if decisions about therapy depend on the result.

  • Tumor marker results should always be interpreted in the context of all available information and the possible influence of other factors (e.g., medication, analytical effects) should be carefully considered.

Use of tumor markers in specific malignancies

International and national guidelines on the clinical management of most cancers are regularly updated (see Table 33.4 ). Modified versions of these guidelines are also frequently adopted and adapted for regional or local use and are similarly readily accessible. Increasing numbers of patients are enrolled in clinical trials with well-defined protocols that may include tumor marker measurements, sometimes as surrogate endpoint indicators.

The optimal use of serum tumor markers for assessing prognosis, monitoring therapy, and detecting recurrence has been studied in greatest detail for choriocarcinoma and GCTs, relatively rare diseases for which tumor marker measurement is mandatory. Serum tumor marker measurements also contribute significantly to the management of more common cancers (e.g., colorectal, ovarian, and prostate) and are less widely used for others (e.g., bladder, breast, and lung). In contrast, measurement of several tissue tumor markers is mandatory for management of breast cancer and is becoming increasingly important in other cancers (e.g., lung and melanoma). The extent to which tumor markers currently contribute to the management of a number of important malignancies is briefly reviewed in the following sections.

Bladder cancer

Approximately 712,000 Americans were living with bladder cancer in 2017, with 81,400 new cases expected to be diagnosed in 2020. Smoking can affect the risk of bladder cancer. The most common symptom of bladder cancer is intermittent hematuria, which is present in 80 to 85% of patients. Presenting symptoms may also include voiding problems or dysuria. Transitional cell carcinomas (TCCs) account for the majority of bladder cancers, but adenocarcinomas, squamous cell carcinomas, and sarcomas also occur. Diagnosis is usually established by cystoscopic evaluation. Cytology of cells shed into the urine is very effective in identifying high-grade bladder cancers but misses many papillary urothelial neoplasms of low malignant potential. The majority of bladder cancer patients are diagnosed with nonmuscle invasive tumors. Primary treatment is complete surgical resection, usually by transurethral resection with or without intravesical treatments with bacille Calmette-Guérin immunotherapy or intravesical chemotherapy. , Radiotherapy may also be required. Even when resection is considered to be complete, there is a high risk of recurrence, and 50 to 70% of patients will develop tumor recurrence within 5 years, depending on the stage of disease. Lifelong surveillance is therefore required.

Tumor markers relevant to bladder cancer.

Markers evaluated for bladder cancer include nuclear matrix proteins (NMPs), human complement factor H-related protein, fibronectin, telomerase, cytokeratins, and survivin. NMPs make up the internal structure of the nucleus and contribute to the regulation of some of the key functions that occur in the nucleus, including DNA replication and RNA synthesis. Some studies suggest that NMPs released by cancer cells may differ from those in normal cells and that different types of cells may have different NMPs.

The FDA has approved an enzyme-linked immunosorbent assay (ELISA) for the measurement of nuclear mitotic apparatus protein (NMP), a component of the nuclear matrix that is overexpressed in bladder cancer. Approved uses of the NMP-22™ test are as an aid in the diagnosis of symptomatic patients or those with risk factors suggesting TCC and in the management and monitoring of patients with TCC. A qualitative point-of-care version of the test is available as an aid in monitoring patients with a history of bladder cancer is also approved by the FDA.

The bladder tumor–associated antigens (BTA), human complement factor H-related protein, and related proteins are involved in the regulation of the alternative pathway of complement activation that prevents complement-mediated damage to healthy cells. It has been suggested that BTA may allow tumor cells to evade the host immune system by preventing tumor cell lysis by immune cells. The BTA-Trak and BTA-Stat tests have been approved by the FDA for use as an aid in conjunction with cystoscopy in the management of bladder cancer patients.

None of the currently available urine tumor markers for bladder cancer are sensitive enough to eliminate the need for cystoscopy, and cytology remains integral to the detection of occult bladder cancer. Urinary biomarkers miss a substantial proportion of patients with bladder cancer and produce false-positive results in others. However, results of an evaluation in which sensitivity and specificity ranges were compared for seven commercially available tumor marker assays suggest that because these tests have relatively high sensitivities, their measurement could be used to extend the period between cystoscopies during surveillance of patients with TCC. Current NCCN recommendations are that use of urinary tumor markers for bladder cancer is optional. Tumor marker use may be considered during surveillance of high-risk nonmuscle invasive bladder cancer but it remains unclear whether these tests provide additional information that is useful for their detection and management.

Breast cancer

Breast cancer is the most common cancer in women worldwide, affecting 10 to 12% of women. In symptomatic women, the main presenting features include a lump in the breast, nipple change, or discharge and skin contour changes. More than 270,000 new cases are likely to be diagnosed in the United States in 2020, with approximately 42,000 deaths and nearly 3 million women living with the disease. Worldwide the incidence appears to be increasing, but in some Western countries, mortality rates are declining, This decrease has been attributed to earlier detection by systematic screening with mammography, greater awareness among women of early signs of breast cancer, and the availability of adjuvant treatment for newly diagnosed cases. While 5-year relative survival rates increased from 75.2% in 1975 to 89.7% in 2003, survival rates have not changed significantly since then. For the approximately 60% of breast cancer patients in the United States who have localized disease at diagnosis (i.e., confined to the primary site), the 5-year survival rate is 98.6%, compared with 25.9% for the 6% of patients who have distant metastases at diagnosis.

Primary treatment for localized breast cancer is either breast-conserving surgery and radiation or mastectomy. , Following primary treatment, most women with invasive breast cancer receive systemic adjuvant therapy such as chemotherapy, hormone therapy, or immunotherapy, or a combination of these. Depending on estrogen receptor (ER) status and other factors, not all patients with breast cancer require adjuvant treatment.

Tumor markers used in the management of breast cancer.

Biomarkers are mandatory for the optimum management of patients with breast cancer. Tissue-based tumor markers that help determine prognosis and guide treatment of invasive breast cancer include those for estrogen receptors, HER2, Ki67, and multigene signatures (see Table 33.7 ). CA15-3 and the closely related BR27.29 (see Table 33.6 ) are the serum markers of choice for breast cancer. Measurement of CEA (see Table 33.5 ) may also be helpful in patients with metastatic breast cancer. In early-stage disease, concentrations of CA15-3 may be similar to those found in healthy women or women with benign breast disease. Emerging blood-based biomarkers such as circulating tumor DNA (ctDNA) and circulating tumor cells (CTC) also show promise in the management of this disease.

Screening and diagnosis.

Early detection undoubtedly improves 5-year survival, but the low specificity and sensitivity of currently available serum tumor markers, especially in early-stage disease, precludes their use in screening for breast cancer. In practice, x-ray imaging with mammography is the only screening modality. Whether the benefits of such screening outweigh the harms associated with overdiagnosis and exposure to radiation is subject to continuing debate. Results of clinical trials conducted over long periods of time may be influenced by confounding factors not originally accounted for during trial design—for example, introduction of more effective therapies.

An independent review commissioned by Cancer Research UK and the English Department of Health concluded in 2013 that the UK breast screening programs confer significant benefit and should continue. Review of older randomized controlled trials and more recent observational studies suggested a 20% reduction in mortality in women invited to screening. This corresponded to one breast cancer death avoided for every 235 women invited to screening, and one death avoided for every 180 women who attend screening. In contrast, authors of a Cochrane review evaluated results of seven trials involving 600,000 women in the age range 39 to 74 years who were randomly assigned to receive screening mammograms or not. The studies with the most reliable information showed that screening did not reduce breast cancer mortality. Assuming screening reduces breast cancer mortality by 15% after 13 years of follow-up, and overdiagnosis and overtreatment is at 30%, the authors concluded that for every 2000 women invited for screening over 10 years, 1 will avoid dying of breast cancer, 10 healthy women who would not have been diagnosed without screening will be treated unnecessarily, and more than 200 women will experience significant ongoing psychologic distress because of false-positive findings. In a complementary study, Surveillance, Epidemiology and End Results (SEER) data were used to examine trends in breast cancer incidence from 1976 through 2008. The authors estimated that in 2008, breast cancer was overdiagnosed in more than 70,000 women in the United States (31% of all breast cancers diagnosed that year) and that despite the substantial increase in early breast cancers detected, screening is having at best only a small effect on the rate of death from breast cancer. The authors cautioned that the question “Should I be screened for breast cancer?” is not answered by their study, but clearly there is a need to inform women considering screening of the advantages and disadvantages. The Cochrane review includes an evidence-based leaflet for laypeople.

Women who are at increased risk of breast cancer because of a strong family history of breast cancer (e.g., relatives diagnosed at a young age) or because they are carriers of the BRCA1 , BRCA2, or TP53 genetic mutations are likely to benefit from additional screening with MRI. This is the current policy in the United Kingdom.

Definitive diagnosis of breast cancer requires biopsy and histology. Serum tumor markers do not contribute to this, but preoperative measurements of CA15-3 and/or CEA are desirable if either marker is going to be used for post-treatment monitoring. A high CA15-3 concentration (e.g., >40 to 50 kU/L) in a patient with apparently localized breast cancer may prompt further investigation to exclude the possibility of metastatic disease.

You're Reading a Preview

Become a Clinical Tree membership for Full access and enjoy Unlimited articles

Become membership

If you are a member. Log in here