Tumor Immunology

Published on 09/02/2015 by admin

Filed under Allergy and Immunology

Last modified 22/04/2025

Print this page

rate 1 star rate 2 star rate 3 star rate 4 star rate 5 star
Your rating: none, Average: 0 (0 votes)

This article have been viewed 2333 times

Tumor Immunology

Oncology is that branch of medicine devoted to the study and treatment of tumors. The term tumor is commonly used to describe a proliferation of cells that produces a mass rather than a reaction or inflammatory condition. Tumors are neoplasms and are described as benign or malignant. Most tumors are of epithelial origin (ectoderm, endoderm, or mesoderm); the remaining tumors are of connective tissue origin (Fig. 33-1). The key distinction between benign and malignant tumors is the ability of malignant tumors to invade normal tissue and metastasize to other secondary sites.

Cancer Stem Cells

Biology research studies have discovered that stem cells are critical for the generation of complex multicellular organisms and the development of tumors. To cure a cancer through stable long-term remission, the stem cell compartment of a tumor needs to be eradicated. Stem cells have three distinctive properties:

If normal self-renewal is subverted, it becomes abnormal self-renewal. If increased self-renewal occurs, combined with the intrinsic growth potential of stem cells, it may yield a malignant phenotype. It is possible that cancer stem cells can arise by mutation from normal stem cells or mutated progenitor cells (Fig. 33-2).

Types of Tumors

Benign Tumors

Benign tumors are often named by adding the suffix -oma to the cell type (e.g., lipoma), but there are exceptions (e.g., lymphomas, melanomas, hepatomas). Benign tumors arising from glands are called adenomas; those from epithelial surfaces are termed polyps or papillomas.

Benign tumors are characterized by the following:

Other types of tumors include non-neoplastic lesions associated with an overgrowth of tissue that is normally present in the organ (e.g., hyperplastic tissue) and choristomas, normal tissue in a foreign location (e.g., pancreatic tissue in the stomach).

Malignant Tumors

A malignant neoplasm of epithelial origin is referred to as carcinoma, or cancer. Those arising from squamous epithelium (e.g., esophagus, lung) are called squamous cell carcinomas, those arising from glandular epithelium (e.g., stomach, colon, pancreas) are called adenocarcinomas, and those arising from transitional epithelium in the urinary system are called transitional cell carcinomas.

Other types of malignant tumors include amine precursor uptake and decarboxylational tumors. These are neuroendocrine tumors that commonly develop from neural crest and neural ectoderm (e.g., small cell carcinoma of lung). Sarcomas, malignant tumors of connective tissue origin (e.g., fibrosarcoma), and teratomas are derived from all three germ cell layers (e.g., teratoma of the ovary or testis).

Malignant tumors are characterized by the following:

Biologically distinct and relatively rare populations of tumor-initiating cells have been identified in cancers of the hematopoietic system, brain, and breast. Cells of this type have the capacity for self-renewal, the potential to develop into any cell in the overall tumor population, and the proliferative ability to drive continued expansion of the population of malignant cells. The properties of these tumor-initiating cells closely parallel the three features that define normal stem cells. Malignant cells with these functional properties are termed cancer stem cells (Fig. 33-3). Cancer stem cells can be the source of all the malignant cells in a primary tumor.

Despite decreases in the incidence of some cancers and associated mortality, cancer remains highly lethal and very common. About 41% of Americans will develop some form of cancer, including nonmelanoma skin cancer, in their lifetime; 20% of Americans will die from cancer. Cancer is the second leading cause of death in the United States.

Epidemiology

Lung, colorectal, and breast cancers are the leading causes of cancer deaths in the United States. The types of cancer that have been increasing in incidence are cancer of the lung, breast, prostate, and pancreas and multiple myeloma, malignant melanoma, and Hodgkin’s lymphoma. The types of cancer that are decreasing in incidence are cancer of the stomach, cervix, and endometrium.

Risk Factors

Risk factors are important in specific cancers. Smoking is responsible for one third of cancers. Other risk factors include a high-fat, low-fiber diet, obesity, and a sedentary lifestyle. Certain types of cancer are more prevalent in specific populations. For example, U.S. blacks have a 20% greater prevalence of cancer than whites. The risk of breast cancer increases with age, and deaths are related to geography. Risk factors for breast cancer include family history, particularly breast cancer in a first-degree relative, first pregnancy after age 30 years, presence of fibrocystic disease, probably the use of oral contraceptives or hormone replacement therapy, prior breast or chest wall radiation, prior breast cancer, and ethanol consumption.

Survivors of childhood and adolescent cancer constitute one of the higher risk populations. The curative therapy (e.g., chemotherapy, radiation) administered for the cancer also affects growing and developing tissues. These patients are at increased risk for early mortality caused by second cancers and cardiac or pulmonary disease. Two thirds of survivors have at least one chronic or late-occurring health problem.

Causative Factors in Human Cancer

Factors that cause most neoplasms are unknown. They can be classified as environmental factors (e.g., chemical and radiation), host factors and disease associations, and viruses.

Environmental Factors

The incidence of cancer has been correlated with certain environmental factors. Table 33-1 lists environmental factors that have been definitively linked with cancer, including aerosol and industrial pollutants, drugs, and infectious agents. Radiation exposure is also known to be associated with specific types of cancer (e.g., acute leukemia, thyroid cancer, sarcomas, breast cancer). Women concerned about organochlorine substances (e.g., polychlorinated biphenyls [PCBs], dioxins, pesticides [DDT, banned in 1972]) can be reassured that available evidence does not suggest an association between exposure to these chemicals and breast cancer.

Table 33-1

Selected Environmental Factors Associated With Cancer

Factor Type of Cancer
Aerosol and Industrial Pollutants
Asbestos (silica) Mesothelioma
Lead, copper, zinc, arsenic, cyclic aromatics, tobacco Lung cancer
Vinyl chloride Liver angiosarcoma
Benzene Leukemia
Aniline dyes, coal Skin and bladder carcinoma
Drugs
Androgenic steroids Hepatocellular carcinoma
Stilbestrol (prenatal) Vaginal adenocarcinoma
Estrogen (postmenopausal) Endometrial carcinoma
Hydantoins Lymphoma
Chloramphenicol, alkylating agents Leukemias, lymphomas
Infectious Agents
Epstein-Barr virus Burkitt’s lymphoma, nasopharyngeal cancer (?)
Hodgkin’s disease
Human papillomavirus Cervical cancer
Herpesvirus type 2 Cervical cancer
Human immunodeficiency virus (HTLV-III) Kaposi’s sarcoma, non-Hodgkin’s lymphoma, primary lymphoma of the brain, bladder cancer
HTLV-I Non-Hodgkin’s lymphoma
Hepatitis B Hepatocellular carcinoma

image

Most chemical carcinogens are inactive in their native state and must be activated by enzymes in the cytochrome P-450 or other enzyme systems (e.g., bacterial enzymes or enzymes induced by alcohol).

In radiation carcinogenesis, ionizing particles (e.g., alpha and beta particles, gamma rays, x-rays) hydrolyze water into free radicals, which are mutagenic to DNA by activating proto-oncogenes. Ultraviolet (UV) light, especially UVB, induces the formation of thymidine dimers, which distort the DNA molecule, leading to skin cancers (e.g., basal cell carcinoma, malignant melanoma).

Host Factors and Disease Associations

Various host factors have been linked to a higher than expected incidence of cancer. For example, the presence of certain genetic disorders (e.g., Down syndrome) is associated with an increased incidence of leukemia. The link between certain genetic abnormalities and leukemia is consistent with a germinal or somatic mutation in a stem cell line.

Familial clustering of germ cell tumors, malignant tumors arising in the testis, has been observed, particularly among siblings. Cryptorchidism and Klinefelter’s syndrome are predisposing factors in the development of germ cell tumors arising from the testis and mediastinum, respectively.

The incidence of cancer is 10,000 times greater than expected in patients with an immunodeficiency syndrome. The increased incidence of lymphomas in congenital, acquired, and drug-induced immunosuppression is consistent with the failure of normal immune mechanisms or antigen overstimulation with a loss of normal feedback control. Table 33-2 lists other cancer-related conditions.

Table 33-2

Cancer-Related Conditions

Disease Related Cancer
Paget’s disease Osteogenic sarcoma
Cryptorchidism Testicular cancer
Neurofibromatosis Brain tumors, sarcoma
Esophageal webbing Esophageal carcinoma
Achlorhydria and pernicious anemia Gastric carcinoma
Cirrhosis Hepatoma
Cholelithiasis Gallbladder cancer
Chronic inflammatory bowel disease Colon cancer
Migratory thrombophlebitis Adenocarcinoma, especially pancreatic
Myasthenia gravis, pure red cell aplasia, T cell disorder Thymoma
Nephrotic syndrome Membranous carcinomas; lymphomas, especially Hodgkin’s

Viruses

Viral causes of some cancers are known. Viruses associated with specific cancers are listed in Table 33-1. Nonpermissive cells that prevent an oncogenic RNA or DNA virus from completing its replication cycle often produce changes in the genome that result in the activation of proto-oncogenes or inactivation of suppressor genes.

Stages of Carcinogenesis

Some precancerous conditions progress through a series of growth alterations before becoming cancerous. For example, cervical cancer progresses from squamous metaplasia to squamous dysplasia to carcinoma in situ, and finally to invasive cancer. Endometrial cancer progresses from endometrial hyperplasia to atypical endometrial hyperplasia to carcinoma in situ, and finally to invasive cancer.

Cancer (Box 33-1) results from a series of genetic alterations that can include the following:

Mutation or overexpression of oncogenes produces proteins that can stimulate uncontrolled cell growth, whereas mutation or deletion of tumor suppressor genes results in the production of nonfunctional proteins that can no longer control cell proliferation. The mutant cell multiplies and the succeeding generations of cells aggregate to form a malignant tumor.

Interleukin-24 (IL-24), initially called MOB-5, is a protein that is usually secreted by immune system cells in response to injury or infection. Research on colon cancer cells has demonstrated that IL-24, in conjunction with its receptors, appears to give a cancer cell the ability to fuel its own growth. The secreted proteins are released from one cell to transmit a signal to grow, migrate, or survive to another cell. These proteins cannot act alone and must act through a receptor or receptors on the receiving cell.

Cancer-Predisposing Genes

Cancer-predisposing genes may act in the following ways:

Relatively few cancer-predisposing genes have been described. An absence of functional alleles at specific loci, however, allows the genesis of the malignant process (Table 33-3). For example, individuals with certain mutations in the gene BRCA2 are at a very high risk (up to 85%) for developing breast cancer and other cancers (e.g., ovarian cancer) because a DNA repair path cannot properly repair ongoing wear and tear to the DNA.

Table 33-3

Tumors Associated With Homozygous Loss of Specific Chromosomal Loci

Tumor Type Chromosomal Linkage
Multiple endocrine neoplasia, type 2 1
Renal cell carcinoma 3
Lung carcinoma 3
Colon carcinoma, familial polyposis 5
Multiple endocrine neoplasia, type 2a 10
Wilms’ tumor, hepatoblastoma, rhabdomyosarcoma 11
Retinoblastoma 13
Ductal breast carcinoma 13
Colon carcinoma 17
Acoustic neuroma, meningioma 22

image

A mutation in a gene thought to be responsible for colon cancer may initially cause it. This gene, APC, normally limits the expression of a protein, survivin. When APC is altered, survivin works overtime and, instead of dying, stem cells in the colon overpopulate, resulting in cancer. Survivin is overexpressed in colon cancer. It prevents programmed cell death, or apoptosis, the process whereby cells normally die. Rather than dying on schedule, cancer cells instead grow out of control. The APC gene controls the amount of survivin by shutting down its production.

Proto-Oncogenes

Proto-oncogenes act as central regulators of the growth in normal cells that code for proteins involved in growth and repair processes in the body. Proteins such as growth factors or transcription factors are necessary for normal growth.

Genetic mutations in proto-oncogenes produce oncogenes. Oncogene activation causes the overexpression of growth-promoting proteins, resulting in hypercellular proliferation and tumorigenesis. Tumor suppressor genes normally counteract proto-oncogenes by encoding proteins that prevent cellular differentiation. When mutations in tumor suppressor genes cause loss of function, the expressed tumor suppressor proteins are no longer able to suppress cellular growth.

For example, the activation of proto-oncogenes (e.g., ras) involved in the growth process or inactivation of suppressor genes (e.g., p53), which keeps growth in check by binding and activating genes that put the brakes on cell division, is responsible for neoplastic transformation of a cell. Defects in the gene for p53 cause about 50% of all cancers.

p53 Protein

The p53 gene (tumor suppressor gene) is located on chromosome 17 and produces a protein that downregulates the cell cycle. A mutation of p53 is associated with an increased incidence of many types of cancer. The p53 tumor suppressor protein is dysfunctional in most human cancers. Even when p53 is not itself mutant, its regulators (e.g., p14ARF, a p53-stabilizing protein) are often altered. The p53 protein is a key responder to various stresses, including DNA damage, hypoxia, and cell cycle aberrations. Specific molecular pathways that activate p53 depend on the nature of the stress and the cell type. Consequently, these determine the specific downstream effectors and cellular response—apoptosis, growth arrest, or senescence.

It is widely believed that the central role of p53 in tumor suppression is to mediate the response to DNA damage. If p53 is missing when damage occurs, cells do not undergo p53-mediated arrest or apoptosis. Cells that have sustained mutations in oncogenes or tumor suppressor genes because of the damage obtain a growth advantage that fuels the development of cancer.

Apparently, DNA damage itself is not the critical event that leads to cancer, as long as the oncogenic stress pathways that activate p53 are intact. For any given cancer type, p53 dysfunction generally correlates with poor treatment response and poor prognosis; therefore, restoration of p53 function is a potential avenue for therapeutic development. Drugs currently being developed will enhance the function of kinases that activate p53 in response to DNA damage.

Role of Oncogenes

The genetic targets of carcinogens are oncogenes. Oncogenes have been associated with various tumor types (e.g., HER-2/neu with breast, kidney, and ovarian cancers). Oncogenes are considered altered versions of normal genes. Over a lifetime, a variety of mutations can convert a normal gene into a malignant oncogene.

Once an oncogene is activated by mutation, it promotes excessive or inappropriate cell proliferation. Oncogenes have been detected in about 15% to 20% of a variety of human tumors and appear to be responsible for specifying many of the malignant traits of these cells. More than 30 distinct oncogenes, some of which are associated with specific tumor types, have been identified (Table 33-4). Each gene has the ability to evoke many of the phenotypes characteristic of cancer cells.

Table 33-4

Some Oncogenes Formed by Somatic Mutation of Normal Genetic Loci

Oncogene Disorder
ab1 Chronic myelogenous leukemia
myc Burkitt’s lymphoma
N-myc Neuroblastoma
EGFR, HER2 Mammary carcinoma
Ras type Wide variety of tumors

EGFR, Epidermal growth factor receptor; HER2, human EGFR-2.

Major classes of oncogene products involved in the normal growth process of cells include the following:

In addition, tumor suppressor genes (antioncogenes) are guardians of unregulated cell growth (e.g., p53, Rb oncogenes).

Mechanisms of Activation

Point mutations, translocations (e.g., t8;142 in Burkitt’s lymphoma) and gene amplification (multiple copies of the gene with overexpression of products) are mechanisms of activation, as follows:

• Overexpression of the c-erbB-2 (HER2/neu) oncogene is noted in up to 34% of patients with invasive ductal breast carcinoma and predicts poor survival.

• Activation of the ras proto-oncogene (point mutation) is associated with about 30% of all human cancers. About 25% of patients with acute myelogenous leukemia display this point mutation. Ras is mutated frequently in colon and pancreatic cancers; it appears that ras activation leads to unregulated expression of IL-24 and its receptors.

• Translocation of the abl proto-oncogene from chromosome 9 to chromosome 22 with formation of a large bcr-abl hybrid gene on chromosome 22 (Philadelphia chromosome) results in chronic myelogenous leukemia.

• Inactivation of suppressor genes (point mutations) leads to unrestricted cell division, inactivation of each of the RB1 suppressor genes on chromosome 13 is associated with malignant retinoblastoma in children, and inactivation of the p53 suppressor gene on chromosome 17 accounts for 25% to 50% of all malignancies involving the colon, breast, lung, and central nervous system.

Tumor-Suppressing Genes

A very different class of cancer genes has been discovered. These tumor-suppressing genes in normal cells appear to regulate the proliferation of cell growth. When this type of gene is inactivated, a block to proliferation is removed and cells begin a program of deregulated growth, or the genetically depleted cell itself may proliferate uncontrollably. Thus, tumor-suppressing genes are referred to as antioncogenes. In time, their discovery will lead to the reformulation of ideas about how the growth of normal cells is regulated.

Much speculation surrounds the operation of tumor-suppressing genes in normal tissue. It is known that normal cells exert a negative growth influence on each other within a tissue. Normal cells also secrete factors that are negative regulators of their own growth and that of adjacent cells. Diffusible factors may also be released by normal cells to induce the end-stage differentiation of other cells in the immediate environment; these factors include the following:

Normal gene products appear to prevent malignant transformation in some way. It is speculated that normal cells must have receptors that detect the presence of these growth-inhibiting and differentiation-inducing factors, which allow them to process the signals of negative growth and respond with appropriate modulation of growth. Genes may specify proteins necessary to detect and respond to the negative regulators of growth. If this process becomes dysfunctional as a result of inactivation or the absence of a critical component, such as the loss of chromosomal loci, a cell may continue to respond to mitogenic stimulation but lose its ability to respond to negative feedback to cease proliferation. Animal experiments have suggested that human beings carry a repertoire of genes, each of which is involved in the negative regulation of the growth of specific cell types. Somatic inactivation of these genes may be involved in the initiation of tumor cell growth or the transformation of benign tumors into malignant ones. Therefore, the somatic inactivation of tumor-suppressing genes may be as important to carcinogenesis as the somatic activation of oncogenes.

Body Defenses Against Cancer

Although there is no single satisfactory explanation for the success of tumors in escaping the immune rejection process, it is believed that early clones of neoplastic cells are eliminated by the immune response. The growth of malignant tumors is primarily determined by the proliferative capacity of the tumor cells and by the ability of these cells to invade host tissues and metastasize to distant sites. It is believed that malignant tumors can evade or overcome the mechanisms of host defenses (Color Plate 18).

Tumor immunity has the following general features:

Host defense mechanisms against tumors are both humoral and cellular. Effector mechanisms include the following:

Tumor Markers

In tumor immunology, a fundamental tenet is that when a normal cell is transformed into a malignant cell, it develops unique antigens not normally present on the mature normal cell. Tumors frequently produce tumor-specific antigens (TSAs) to which the host may develop antibodies. Virus-induced cancers are the most antigenic; chemical-induced cancers are the least antigenic.

Tumor markers are substances present in or produced by tumors that can be used to detect the presence of cancer based on their measurement in blood, body fluids, cells, or tissue (Table 33-5). A tumor marker may be produced by the host in response to a tumor that can be used to differentiate a tumor from normal tissue or to determine the presence of a tumor. Non-neoplastic conditions can also exhibit tumor marker activity (Table 33-6). Some tumor markers are used to screen for cancer, but markers are more often used to monitor recurrence of cancer or determine the degree of tumor burden in the patient. To be of any practical use, the tumor marker must be able to reveal the presence of the tumor while it is still susceptible to destructive treatment by surgical or other means. Tumor markers can be measured quantitatively in tissues and body fluids using biochemical, immunochemical, or molecular tests (Table 33-7).

Table 33-5

Cancer Biomarkers

Type of Molecule Biomarkers in Blood or Body Fluid Type of Cancer Detected
Enzyme Prostate-specific antigen (PSA) Prostate
Oncofetal proteins Alpha-fetoprotein (AFP); carcinoembryonic antigen (CEA) Hepatocellular, germ cell
Colorectal
Hormones β-Human chorionic gonadotropin (β-hCG); calcitonin; adrenocorticotropic hormone (ACTH) Trophoblastic
Medullary thyroid
Small cell lung
Mucins CA 125, CA 19-9, CA 27.29, CA 15-3 Ovarian
Breast
Immunoglobulins Bence-Jones protein (urine) Multiple myeloma
Genetic alteration HER2/neu Breast
Other proteins HE4 Ovarian
  Tg Thyroid
  Nuclear matrix protein 22 (NMP-22); bladder tumor–associated antigen (BTA)/complement factor H–related protein (CFHrp) Bladder

Adapted from Snyder J: Genomic, proteomic developments in tumor markers, Adv Med Lab Prof 16:42–48, 2004; and Rhea JM, Molinaro RJ: Cancer biomarkers, MLO Med Lab Observer, 43:10–18, 2011.

Table 33-6

Non-neoplastic Conditions With Elevated Serum and Plasma Concentrations of Tumor Markers

Tumor Marker Concentration in Normal Serum (ng/mL) Non-neoplastic Conditions
CEA <2.5 Inflammatory bowel disease, pancreatitis, gastritis, smoker’s chronic bronchitis, alcoholic liver disease, hepatitis
AFP <40 Pregnancy, regenerating liver tissue after viral hepatitis, chemically induced liver necrosis, partial hepatectomy, cystic fibrosis, ataxia-telangiectasia, premature infants, tyrosinemia
β-hCG Negative Pregnancy
Serum acid phosphatase Negative Pregnancy
Placental alkaline phosphatase Negative Pregnancy

Table 33-7

Tumor Markers in Neoplasms

Tumor Markers Clinical Value
CEA Monitors response to therapy of patients with various types of cancer
AFP Diagnosis of germ cell and hepatic tumors
CA 125 Diagnosis of ovarian cancer
β-hCG Diagnosis of germ cell tumors
Prostate acid phosphatase Diagnosis of prostate cancer

AFP, Alpha1-fetoprotein; β-hCG, beta subunit of chorionic gonadotropin; CEA, carcinoembryonic antigen.

The search for tumor markers goes back more than 150 years. The earliest identified tumor marker was Bence-Jones protein, a light-chain immunoglobulin, found in patients with multiple myeloma (see Chapter 27). Over the last 15 years, the use of tumor markers in the United States has risen dramatically. Tumor markers play an especially important role in the diagnosis and monitoring of patients with prostate, breast, and bladder cancers.

Older, well-established markers include alkaline phosphatase and collagen-type markers in bone cancer, immunoglobulins in myeloma, catecholamines and their derivatives in neuroblastoma and pheochromocytoma, and serotonin metabolites in carcinoid. In addition, there are many breast tissue prognostic markers (e.g., hormone receptors, cathepsin-D, HER2/neu oncogenes, plasminogen receptors and inhibitors).

The list of tumor markers approved by the U.S. Food and Drug Administration (FDA) continues to grow (Table 33-8). Nine of these biomarkers are protein biomarkers identifiable in blood. Other recently approved protein biomarkers can be detected in urine, such as nuclear matrix protein 22, fibrin and fibrinogen degradation products, and bladder tumor antigen for monitoring bladder cancer, and by immunohistochemical methods using tumor tissues, such as estrogen receptor for breast cancer. Additional FDA-approved cancer biomarkers are DNA-based, such as human epidermal growth factor receptor 2 and HER2/neu for breast cancer, and can be assayed by fluorescent in situ hybridization (FISH). Multiple-marker combinations are useful in the management of some cancers (Table 33-9), but the use of more than two markers is questionable.

Table 33-8

Some Common Serum Tumour Markers and Their Clinical Utility

Tumor Type Cancer Deaths (USA) (%) Tumour Markers Specificity Sensitivity Tumour Detection Clinical Utility
Lung + bronchus 28 Neuron specific enolase Poor Poor Late Poor
Colon + rectum 9 Carcinoembryonic antigen (CEA) Poor Modest Late Modest
Breast 7 AA 15-3; CEA Poor Modest Late Modest
Pancreas 6 CA 19-9: CEA Poor Poor Late Poor
Prostate 5 Prostate-specific antigen Modest Good Good Good
Stomach 2 CEA; CA 19-9 Modest Modest Late Poor
Ovary 2.5 CA 125 Modest Modest Intermediate Good
Liver 3 Alpha-feto-protein α-FP Good Good Intermediate Good
Myeloma 1.9 Monoclonal protein/FLC Good Good Early Very good
AL amylidosis 0.3 Monoclonal protein/FLC Good Good Early Very good
Germ cell 0.1 α-FP; human chorionic gonadotrophin (HCG) Good Good Early Very good
Choriocarcinoma <0.1 HCG Good Good Early Very good
Neuroendocrine <0.1 Chromogranin, A, gastrin Modest Good Early Very good

image

FLC, Free light chains.

All of these are measured using highly sensitive immunoassays apart from monoclonal proteins.

From Bradwell AR: Serum free light chain analysis, Birmingham, UK, 2010, Binding Site, p 2.

Table 33-9

Related Multiple Tumor Markers

Markers Comments
AFP and β-hCG Valuable combination in therapy and follow-up in patients with germ cell tumors of the testes.
CEA, AFP, and LDH Combination seems to help differentiate primary liver cancer from liver metastases related to another organ.
Ratio of free to total PSA The ratio may distinguish benign prostatic hypertrophy (BPH) from prostate cancer.
CEA and numerous mucin-type markers May complement each other

AFP, Alpha-fetoprotein; β-hCG, beta subunit of chorionic gonadotropin; CEA, carcinoembryonic antigen; LDH, lactate dehydrogenase; PSA, prostate-specific antigen.

An ideal tumor marker would be an assay in which a positive result would only occur in patients with a malignancy, would correlate with stage and response to treatment, and is easily reproducible. No tumor marker to date has met this ideal marker description, nor has any tumor marker has been established as a practical screening test in a general healthy population or in most high-risk populations. The rationale for this poor predictive value of tumor markers is the lack of sensitivity and specificity in the low cancer rates that prevail in population groups. Because of the low prevalence of cancer, in general, even assays that are highly sensitive and specific may have a low predictive value.

Categories of Tumor Antigens

Tumor cells manifest tumor antigens, as well as self HLA antigens. There are four types of tumor antigens identified:

Specific Tumor Markers

Ten protein cancer biomarkers have been FDA-approved for clinical use:

Other markers include the beta subunit of human chorionic gonadotropin (β-hCG) and miscellaneous enzyme and hormone markers.

Alpha-Fetoprotein

Alpha-fetoprotein (AFP) is normally synthesized by the fetal liver and yolk sac. AFP is secreted in the serum in nanogram to milligram quantities in hepatocarcinoma, endodermal sinus tumors, nonseminomatous germ cell (testicular) cancer, teratocarcinoma of the testis or ovary, and malignant tumors of the mediastinum and sacrococcyx. In addition, a small percentage of patients with gastric and pancreatic cancer with liver metastasis may have elevated AFP levels. Both AFP and β-hCG should be quantitated initially in all patients with teratocarcinoma because one or both markers may be secreted in 85% of patients. The concentration of AFP may be elevated in nonneoplastic conditions such as hepatitis and cystic fibrosis.

AFP is a reliable marker for following a patient’s response to chemotherapy and radiation therapy. Levels should be obtained every 2 to 4 weeks (metabolic half-life in vivo, 4 days).

Prostate-Specific Antigen and Prostatic Acid Phosphatase

Prostate cancer is a leading cause of cancer death in U.S. men. Although there has been controversy in recent years about the application of prostate assays, there are two tumor markers for cancer of the prostate, prostate-specific antigen (PSA) and prostatic acid phosphatase.

Prostate-Specific Antigen

PSA screening has been controversial in recent years. Research on PSA testing offers mixed results on the benefits of PSA screening testing. However, some investigators suggest that PSA-screened men were more likely to be treated for prostate cancer at academic centers, where they got more state-of-the-art treatment.

PSA is a prostate tissue–specific marker, but not a prostate cancer–specific marker. It is a protease enzyme secreted almost exclusively by prostatic epithelial cells. Blood levels of PSA are increased when normal glandular structure is disrupted by benign or malignant tumor inflammation. The serum PSA level is directly proportional to tumor volume, with a greater increase per unit volume of cancer compared with benign hyperplasia. However, elevated PSA levels can be detected in prostate infection, irritation, benign prostatic hypertrophy, and recent ejaculation.

Free PSA assists in distinguishing cancer of the prostate from benign prostatic hypertrophy (BPH). Comparison of free PSA to PSA levels is used to assess the risk of cancer because the ratio of free PSA to PSA in prostate cancer is decreased. PSA levels appear useful for monitoring progression and response to treatment in patients with prostate cancer.

Other techniques that have been used for the detection of prostate cancer include PSA velocity (incremental increase of PSA over time), PSA density (ratio of serum PSA to prostate volume), age-adjusted PSA (PSA increases with age), biostatistically derived algorithms, free and total PSA, complexed PSA and, most recently, human kallikrein II, a molecule similar but not identical to PSA.

Carcinoembryonic Antigen

The cell surface protein carcinoembryonic antigen (CEA) is found predominantly on normal fetal endocrine tissues in the second trimester of gestation. If CEA is detected in mature individuals, it is of limited diagnostic value but is helpful in differentiating between benign and malignant pleural and ascites effusions. CEA was first described in 1965 as a tumor marker specifically elevated in patients with colon cancer; it was later found to be elevated in patients with breast, lung, liver, and pancreatic cancers. Plasma levels higher than 12 ng/mL are strongly correlated with malignancy. Elevated neoplastic states frequently associated with an increased CEA level are endodermally derived gastrointestinal neoplasms and neck and breast carcinomas. Also, 20% of smokers and 7% of former smokers have elevated CEA levels.

CEA is used clinically to monitor tumor progress in patients who have diagnosed cancer with a high blood CEA level. If treatment leads to a decline to normal levels (<2.5 ng/mL), a rise in CEA level may indicate cancer recurrence to the clinician. A persistent elevation is indicative of residual disease or poor therapeutic response. In patients who have undergone colon cancer resection surgery, the rate of clearance of CEA levels usually return to normal within 1 month, but may take as long as 4 months. Blood specimens should be obtained 2 to 4 weeks apart to detect a trend.

CA 15-3

CA 15-3 is a biomarker used in conjunction with patient history, physical examination, and mammography during active cancer therapy to monitor metastasis. CA 15-3 is a high-molecular-weight (HMW) glycoprotein coded by the MUC-II gene and expressed on the ductal cell surface of most glandular epithelial cells. The main purpose of the assay is to monitor patients after mastectomy. Using a cutoff of 25 U/mL for CA 15-3, the detection rate is only 5% for stage I breast cancer.

The sensitivity is much better in higher stage disease, which makes it a good measure of tumor burden. CA 15-3 is positive in other conditions, including liver disease, some inflammatory conditions, and other carcinomas. A change in the CA 15-3 concentration is more predictive than the absolute concentration. Over time, tumor markers exhibit a steady state in the body, a balance between antigen production by the tumor and degradation and excretion. Changes in tumor burden are reflected by changes in the tumor marker concentration.

A high CA 15-3 level (>32 U/mL) usually indicates advanced breast cancer and a large tumor burden. This biomarker lacks sensitivity and specificity and is approved only for monitoring patient response to treatment and recurrence.

CA 27.29: Breast Carcinoma–Associated Antigen

Carcinoma of the breast often produces mucinous antigens that are HMW glycoproteins with O-linked oligosaccharide chains. Monoclonal antibodies (MAbs) directed against breast carcinoma–associated antigen (CA 27.29) can quantitate the levels of this antigen in serum. The antibodies recognize epitopes of a breast cancer–associated antigen encoded by the human MUC1 gene, which is also referred to as MAM6, milk mucin antigen, CA 27.29, and CA 15-3. This tumor marker may be useful in conjunction with other clinical methods for predicting early recurrence of breast cancer. It is not recommended as a breast cancer screening assay. Increased levels of CA 27.29 (>38 U/mL) may indicate recurrent disease in a woman with treated breast carcinoma and may indicate the need for additional testing or procedures. Some clinical investigators do not endorse the routine use of this new marker.

Other Cancer Biomarkers

β-Human Chorionic Gonadotropin (β-Beta Subunit)

β-hCG, an ectopic protein, is a sensitive tumor marker with a metabolic half-life in vivo of 16 hours. A serum level of β-hCG higher than 1 ng/mL is strongly suggestive of pregnancy or a malignant tumor such as an endodermal sinus tumor, teratocarcinoma, choriocarcinoma, molar pregnancy, testicular embryonal carcinoma, or oat cell carcinoma of the lung.

Miscellaneous Enzyme Markers

Lactic dehydrogenase (LDH) is a frequently measured enzyme of the glycolytic pathway. The level of LDH is elevated in a wide variety of malignancies and other medical disorders. Its level has been shown to correlate to tumor mass in solid tumors so it can be used to monitor progression of these tumors.

Neuron-specific enolase is an isoenzyme specific for all tumor cells derived from the neural crest. An enzyme increase has been detected in neuroblastoma, pheochromocytoma, oat cell carcinomas, medullary thyroid and C cell parathyroid carcinomas, and other neural crest–derived cancers. Serum levels are frequently elevated in disseminated disease.

Placental alkaline phosphatase (ALP) can be detected during pregnancy. ALP is also associated with the neoplastic conditions of seminoma and ovarian cancer.

Miscellaneous Hormone Markers

Elevated or inappropriate serum levels of hormones can function as tumor markers. Adrenocorticotropic hormone (ACTH), calcitonin, and catecholamines may be secreted by differentiated tumors of endocrine organs and squamous cell lung tumors. Oat cell carcinomas may produce β-hCG, antidiuretic hormone (ADH), serotonin, calcitonin, parathyroid hormone (PTH), and ACTH. These hormones can be used to follow a patient’s response to therapy.

In addition, some breast cancers demonstrate progesterone and estradiol (estrogen) receptors, which are strongly correlated with a positive response to antihormone therapy. Patients with neuroblastoma and pheochromocytoma secrete catecholamine metabolites that can be detected in the urine. Neuroblastomas also release neuron-specific enolase and ferritin; these markers can be used for diagnosis and prognosis.

Breast, Ovarian, and Cervical Cancer Markers

For more than 15 years, circulating breast cancer antigens have been used to monitor therapy and evaluate recurrence of the cancer. Estrogen and progesterone receptors are universally accepted as prognostic markers and therapeutic choice indicators. A relatively new approach has been the use of the oncogene HER2/neu as a prognostic indicator and a marker related to the choice of therapy. This has been particularly useful since the introduction of trastuzamab as a chemotherapeutic agent that targets the HER2/neu receptor. Breast cancer patients who express HER2/neu in their cancers have a poor prognosis with shorter disease-free and overall survival than patients who do not express HER2/neu. The evaluation of HER2/neu has two clinical functions: (1) predictive marker for response to trastuzumab therapy; and (2) prognostic marker.

A newer and more powerful predictor of the outcome of primary breast cancer in young women has been reported. Microarray analysis of a previously established 70-gene profile has demonstrated that a good prognosis gene expression signature is a strongly independent factor in predicting disease outcome.

Epidermal Growth Factor Receptor

EGFR and human epidermal growth factor receptor-2 (HER-2, HER2/neu, or c-erB-2) are both transmembrane tyrosine kinase receptors expressed on normal epithelial cells but overexpressed in some cancer cells. A portion of both receptors is released from the cell surface and circulates in normal people and in abnormally high levels in cancer patients. The shed portions can be measured in serum or plasma using antibody-based immunoassays. These assays allow real-time assessment of the patient’s HER2/neu or EGFR status and repeat testing for patient monitoring; they can be performed in a standardized and quantitative manner.

HER2 and EGFR have been the targets of considerable pharmaceutical activity to develop therapies that will interfere with the oncogenic potential of these growth factor receptors. These therapies include small-molecule inhibitors designed to target and block the function of HER2 protein overexpression. One drug, trastuzumab, is a humanized antibody that targets cells that overexpress the HER2/neu and has been successfully used in combination with chemotherapy to increase the efficacy of the antibody-based treatment. An anti-EGFR antibody known as IMC-225 is directed against cells that overexpress the EGFR oncoprotein.

Molecular Diagnosis of Breast Cancer

The assessment of DNA content (aneuploid, diploid) and cell cycle analysis (G0G1, S, G2, M) can be of prognostic use in certain solid tumors (e.g., breast cancer). Cell cycle analysis can be performed on fresh or frozen tissue. In breast cancer, research has indicated that low S phase and diploid DNA content are associated with a relatively good prognosis; a high S phase number of cells and aneuploid DNA content have a tendency to indicate a worse prognosis. The DNA content of a tumor is classified in order of worsening prognosis from diploid, near-diploid, tetraploid, aneuploid, hypertetraploid, and hypoploid. The ratio of tumor G0G1 DNA content to normal G0G1 DNA content is called the DNA index. Ploidy status and the S phase fraction should be combined with other indicators (e.g., hormone receptor status) to evaluate treatment options and prognosis.

In June 2011, the FDA approved the Inform Dual ISH, a genetic test developed by a Roche affiliate (Ventana Medical Systems, Tucson, Ariz). This test helps determine whether breast cancer patients are HER2-positive, which makes them candidates for trastuzumab therapy. The Dual ISH test was designed to detect amplification quantitatively by light microscopy of the HER2 gene using two-color chromogenic in situ hybridization (ISH) in formal-fixed, paraffin-embedded human breast and gastric cancer. An advantage of this procedure is that it is possible to view HER2 and chromosome 17 signals directly under a microscope and for a longer period.

Bladder Cancer

Bladder cancer tumor markers for the management of patients with bladder cancer have been actively investigated. Assays approved for clinical use include the following:

Almost all human tumors contain telomerase, a growth enzyme that promotes the malignant proliferation of cancer. Normal cells usually do not have this enzyme, but telomerase renews the DNA of tumor cells and permits indefinite replication.

Telomerase was first observed in ovarian cancer cells and its presence was later established in almost all cancers. It is not clear whether other vital cells need telomerase to function. For example, telomerase inhibition could adversely affect stem cells, which help produce blood cells and lymphocytes and may need the enzyme to function. Second, telomerase inhibition has not been proved or tested physiologically in human beings. Finally, a drug based on telomerase would have to reduce the ability of the cancer to spread. Screening for telomerase inhibitors and plans for future studies to discover and develop chemicals that block the action of telomerase may suggest a design of more effective anticancer drugs.

DNA Microarray Technology

New developments in molecular genetics involve DNA microarray technology (see Chapter 14). Cancer can arise not only from mutations in oncogenes and tumor suppressor genes, but also from genes involved in cell cycle control, DNA repair, and apoptosis. Microarrays have the potential to uncover signature gene expression patterns for specific cancers and ultimately assist in the staging of tumors, prognosis, and treatment. Microarrays may help disclose global gene expression pattern differences between healthy and diseased cells as more sensitive and specific diagnostic markers are developed, such as CD44+/CD24− gene expression profile in breast cancer versus normal breast tissue. When differentially expressed genes were used to generate a 186-gene invasiveness gene signature (IGS), the IGS was strongly associated with metastasis-free survival and overall survival for four different types of tumors.

Proteomic technology uses two-dimensional polyacrylamide gel electrophoresis (2D-PAGE) and mass spectrometry. Although these techniques are not revolutionary, advances have improved their sensitivity. Expansion of computer-assisted bioinformatics has simplified the process of protein identification from mass spectra. Mass spectra are proving to be comparable to CA 125 for the detection of early-stage ovarian cancer.

In colorectal cancer, fecal DNA screening has been demonstrated to be useful. Oncogene mutations that characterize colorectal neoplasia are detectable in exfoliated epithelial cells in the stool. Neoplastic bleeding is intermittent but epithelial shedding is continual, potentially making fecal DNA testing more sensitive.

What’s New in Cancer Diagnostic Testing?

Next Generation Sequencing (NGS)

Next Generation Sequencing (NGS) as described in Chapter 14, Molecular Techniques, is another step toward personalized cancer treatment. Three aspects of importance in NGS are:

Continuous Field-Flow Assisted Dielectropheresis (DEP)

The ability to isolate and characterize rare circulating tumor cells (CTCs) may provide critical insights into primary tumors, the process of metastasis, and monitor disease progression. Performing molecular analysis of CTCs offers a unique approach for genotyping patient-specific tumors and mutations as well as guiding treatment options.

To date, only one technology is FDA approved for use with only three tumor types: prostate, breast, and colorectal cancers. The new technology is antibody-dependent, that means the detection and capture of CTCs depends on antigen expression of the surface of cancer cells of epithelial origin, e.g., EpCAM. A new, next-generation antibody-independent technology has recently been developed. It relies on continuous field-flow assisted dielectropheresis (DEP) to isolate and recover CTCs from the blood of cancer patients. This technology has already proven to be successful in detecting and isolating a wider range of cancers in greater cells quantities, and research protypes are now being used in phase I, phase II, and phase III clinical studies. The isolation of rare cells from blood using DEP field-flow assist is based on the differences in dielectric properties between bloods, e.g., lymphocytes, monocytes, and granulocytes, and solid tissue-deprived cancer cells. This technology is revolutionary because:

Future applications of this technology are being explored to facilitate implementation of personalized medicine with improved clinical outcomes.

Modalities For Treating Cancer

Many different modes of therapy, including angiogenesis inhibitors, which keep tumors from building new blood vessels to supply themselves with food and oxygen, have demonstrated effectiveness in the treatment of cancer (Table 33-10).

Table 33-10

Immunotherapy in Malignant Disease

Approach Agent Proposed Mechanism
Active
Specific Modified or unmodified tumor cells, cell extract Cellular and/or humoral response
Nonspecific systemic Bacille Calmette-Guérin (BCG); methanol-extracted residue of mycobacterial skeletal wall, Corynebacterium parvum, Pseudomonas vaccine; levamisole, interferon General immunocompetence; increased mononuclear phagocyte system activity; restores immunocompetence
Local BCG; virus, hapten, dinitrochlorobenzene Macrophage activation; killing of tumor with bystander effect
Passive
Adoptive specific Allogeneic organogenesis antibody; targeted monoclonal antibody; lymphocytes, lymphocyte extract (immune RNA transfer factor); lymphokine-activated killer cells Removes soluble antigen or directly kills target cell; conjugated with antitumor drug or radioisotope; transfer of immunity; cytolysis of tumor cells

image

Chemotherapeutic Agents

Drugs are used in cancer therapy for cure, palliation, and research to develop more effective therapy. The mechanisms of drug action are linked to the mitotic cell cycle; thus, antitumor drugs may be placed in the following three classes:

Cell Cycle Active, Phase Specific

Drugs in the cell cycle active, phase-specific category act on the S, G2, or M phase of mitosis. S phase–active drugs are divided into antimetabolites, antifolates, and synthetic enzyme inhibitors. Antimetabolites act through the incorporation of a nucleotide analogue into DNA, resulting in an abnormal nucleic acid (e.g., 5-fluorouracil, 6-mercaptopurine, 6-thioguanine, fludarabine). The antifols act as competitive inhibitors of the enzyme dihydrofolate reductase, which is necessary for the generation of CH3 groups required for thymidine synthesis (e.g., methotrexate). Synthetic enzyme inhibitors include DNA polymerase inhibitor (cytosine arabinoside) and nucleotide reductase inhibitor (hydroxyurea).

G2 phase active drugs include bleomycin, which is thought to cause fragmentation of DNA, and etoposide (Eposin, Etopophos, VePesid, VP-16), which is thought to cause double-stranded breaks in DNA by complexing with topoisomerase.

M phase active drugs include vinca alkaloids (e.g., vincristine, vinblastine), which are thought to inhibit the mitotic spindle apparatus, and paclitaxel (Taxol), which stabilizes microtubules.

Non–Cell Cycle Active

Drugs in the non–cell cycle active category can be divided into five types—alkylating agents, l-asparaginase, corticosteroids, hormone antagonists, and miscellaneous. Alkylating agents (e.g., nitrogen mustard and mustard derivatives—mechlorethamine [Mustargen], cyclophosphamide [Cytoxan], chlorambucil [Leukeran], and melphalan [Alkeran]) act by interstrand cross-linking of DNA, thereby preventing normal DNA replication. This interference is not only cytotoxic, but also potentially mutagenic and carcinogenic. l-Asparaginase inhibits protein synthesis.

Glucocorticosteroids are the most frequently used steroids. Steroids control the damaging inflammatory immune response. The target cells are monocytes and T lymphocytes. Monocytes block IL-1 production, block TNF-γ, and reduce chemotaxis. The consequences are inhibition of T cell activation, activation and recruitment of monocytes and neutrophils, and inhibition of the migration of cells to the site of inflammation. The steroids used in cancer oncology include glucocorticoids (prednisone), estrogens (diethylstilbestrol), androgens (testosterone propionate), and progestational agents (medroxyprogesterone, megestrol acetate).

Hormone estrogen antagonists (e.g., tamoxifen) competitively bind to specific cytoplasmic receptors.

Cytokines

Cytokines constitute another group of cancer chemotherapy drugs (see Chapter 5). IFN, IL-2, and colony-stimulating factors (CSFs) have been used to treat certain types of cancer in patients. Currently, IFNs are used to treat patients with hairy cell leukemia, chromic myelogenous leukemia, and multiple myeloma. IL-2 is used in the treatment of renal cell carcinoma and melanoma. CSFs decrease the duration of chemotherapy-induced neutropenia and may permit more dose-intensive therapy.

Effects of Drug-Induced Immunosuppression

Drugs used to treat malignancies such as solid tumors or leukemia can have profoundly suppressive effects on the inflammatory response, delayed hypersensitivity, and specific antibody production (Table 33-11). Examples of the immune depression induced by drugs include depletion of T cells by corticosteroids, caused by the blocking of egress from the bone marrow into the circulation, and dysfunction of the antibody response, caused by folate antagonists and purine analogues. Thus, infection secondary to immune suppression is a major cause of death in cancer patients beginning therapy and those who are in clinical remission.

Table 33-11

Effects of Chemotherapy on the Immune Response

Chemotherapeutic Agent Antibody Delayed Hypersensitivity
Primary Response Secondary Response Primary Response (Initial) Secondary Response (Recall)
Corticosteroid 0 0 + + +
Methotrexate + + + + 0
6-Mercaptopurine 0 + + 0
Azathioprine 0 + + 0
6-Thioguanine 0 + + 0
Cytosine arabinoside + + + 0 0
Cyclophosphamide + + 0 + 0
l-Asparaginase + 0 0 0
Daunomycin + 0 + 0

image

Recent Advances

Monoclonal antibody (MAb) technology began with the winning contribution of Köhler, Milstein, and Jerne, who won the Nobel Prize in Physiology or Medicine in 1984. This led to great expectation that MAbs would provide effective targeted therapy for cancer. After early enthusiasm for MAbs, clinical trials were disappointing in the 1980s and early 1990s with one exception, antiidiotype antibodies in follicular lymphoma. When success was finally observed in hematologic malignancies, the importance of the antigen target specificity and developing humanized MAbs was recognized. The major success of mAB therapy has been seen with anti-CD20 MAbs. Anti-CD20 rituximab (Table 33-12) was the first MAb to be approved by FDA for use in relapsed indolent lymphoma. Today, rituximab is widely accepted to be the single most important factor leading to improved prognosis in a range of B cell lymphomas and, more recently, in B cell chronic lymphocytic leukemia (CLL). However, some patients develop resistance to rituximab, which provides a challenge for research.

Table 33-12

Current FDA-Approved Antibodies for Cancer Treatment

Generic Name Trade Name Composition Antigen Target Treatment
Applications
Alemtuzumab Campath Humanized IgG1 monoclonal antibody with murine binding region for CD52 CD52 Chronic lymphocytic leukemia
Bevacizumab Avastin Humanized IgG1 monoclonal antibody with murine binding region for vascular endothelial growth factor Vascular endothelial growth factor Prevention of vascularity in tumor antiangiogenesis (e.g., colorectal and nonsmall cell lung cancers)
Cetuximab Erbitux Chimeric monoclonal-murine Fab variable portion and human IgG1 kappa Fc portion Epidermal growth factor receptor Colorectal, head and neck cancers
Rituximab Rituxan Chimeric monoclonal-murine Fab variable portion and human IgG1 kappa Fc portion CD 20 on B-lymphocytes Non-Hodgkin’s lymphoma
Trastuzumab Herceptin Humanized IgG1 kappa monoclonal with murine binding region for HER2 HER2/neu Breast cancer

image

Immunotherapy for tumors can take the form of active or passive therapy. Active host immune responses may be achieved by the following:

• Vaccination with killed tumor cells or with tumor antigens or peptides. New research studies have suggested that anti-CD20 MAb may induce an adaptive antitumor immune response or vaccination effect, which may underlie the durable remissions experienced by some patients after anti-CD20 MAb treatment.

• Enhancement of cell-mediated immunity to tumors by expressing costimulators and cytokines and treating with cytokines that stimulate the proliferation and differentiation of T lymphocytes and NK cells.

• Nonspecific stimulation of the immune system by the local administration of inflammatory substances or by systemic treatment with agents that function as polyclonal activators of lymphocytes.

• For the first time in the history of cancer treatment, gene therapy has apparently succeeded in shrinking and even eradicating large metastatic tumors. Inserting genes into a patient’s cells enables the body to fight a disease on its own, without medication.

Passive immunotherapy consists of the following:

What’s New in Drug Therapy?

The development of inhibitors to target proteins encoded by mutated cancer genes has now been achieved, with repeated success. The first victory was imatinib (Gleevec), approved by the FDA in 2001, a potent inhibitor of the Abelson (ABL) kinase in chronic myeloid leukemia (CML).

This is an important example of therapeutic targeting of the products of genomic alterations in a specific cancer. After the referencing of the genome sequence, cancer genomes have been identified for several new mutated cancer genes. Unfortunately, many mutated cancer genes do not make tractable targets for new drug development. The International Cancer Genome Consortium and the Cancer Genome Atlas are using next-generation sequencing technologies for tumors from 50 different cancer types to generate more than 25,000 genomes at genomic, epigenomic, and transcriptomic levels. This should generate a complete catalogue of oncogenic mutations, some of which may prove to be new therapeutic targets.

The list of drugs used for cancer therapy continues to grow. The new therapeutic agents target various modes of action and applications (Table 33-13).

Table 33-13

Targeted Therapeutic Agents in Cancer

Classification Gene Genetic
Alteration
Drug Application
Nonreceptor tyrosine kinase ABL Translocation
(BCR-ABL)
Imatinib Chronic myelogenous leukemia
Receptor, tyrosine kinase ECFR Mutation, amplification Gefitinib, erlotinib Lung cancer, glioblastoma
Serine-threonine-lipid kinase PI3K PIK3CA mutations BEZ235 Colorectal, breast, gastric cancer, glioblastoma
DNA damage or repair BRCA1 and
BRCA2
Mutation
(synthetic lethal effect)
Olaparib, MK-4827 (PARP inhibitor) Breast, ovarian cancer

image

Adapted from McDermott U, Downing JR, Stratton MR: Genomics and the continuum of cancer care, N Engl J Med 364:340–350, 2011.

CASE STUDY 2

MS, a 65-year-old black woman, visited her primary care provider for an annual examination, including a routine pelvic examination. Although she had gained some weight since her last examination, she reported that her general health was good but that she had been experiencing some gastrointestinal problems over the last 6 weeks.

A palpable mass was discovered during her pelvic examination. A CA 125 assay and a transvaginal ultrasound examination were ordered.

The patient’s CA 125 was 425 U/mL (reference range, <35 U/mL). The presence of a mass in the right side of the abdomen and abdominal ascites were confirmed.

The patient had a total abdominal hysterectomy with a bilateral salpingo-oophorectomy; 4 weeks after operation, she began a chemotherapy series. The patient was judged to be in remission for 6 months when recurrence of the tumor was noted with diagnostic imaging. Subsequent chemotherapy was ineffective, and the patient died 8 months later.

Question

See Appendix A for the answers to multiple choice questions.

image Prostate-Specific Antigen (PSA) Rapid Test of Seminal Fluid (SeraTEc, Goettingen, Germany)

Principle

The test is a chromatographic immunoassay (CIA) for the rapid semi-quantitative determination of PSA in body fluids. It contains two monoclonal murine anti-PSA antibodies as active compounds. One of these antibodies is immobilized at the test region on the membrane. A glass fiber pad downstream of the membrane is used for sample loading and transmission to a second fiber pad with the dried and gold labeled second monoclonal murine anti-PSA antibody. PSA at the sample will bind to the remobilized gold-labeled antibody and form a PSA-gold-labeled-anti-PSA-antibody-complex. The colored gold labeled anti-PSA-antibody will bind to the anti-mouse antibody at the control region and the region of the internal standard thus developing two red lines (one at the control region and one at the region of the internal standard). If the sample contains PSA, the PSA-gold-labeled anti-PSA-antibody complex will bind to the immobilized monoclonal antibody of the test result region that recognizes another epitope on the PSA molecule (sandwich complex). The binding is indicated by the formation of an additional line.

Chapter Highlights

• Tumors are neoplasms described as benign or malignant. A benign neoplasm is a nonspreading tumor; a malignant neoplasm is a growth that infiltrates tissues, metastasizes, and often recurs after attempts to remove it surgically.

• A malignant neoplasm can be referred to as carcinoma or cancer.

• The incidence of cancer has been correlated with certain environmental factors (e.g., occupational exposure to known carcinogenic agents) and host susceptibility.

• Cancer often begins when a carcinogenic agent damages the DNA of a critical gene in a cell. The mutant cell multiplies and the succeeding generations of cells aggregate to form a malignant tumor.

• Proto-oncogenes act as central regulators of the growth in normal cells and are antecedents of oncogenes.

• The genetic targets of carcinogens, oncogenes, have been associated with various tumor types, largely from preexisting genes present in the normal human genome. Oncogenes are considered altered versions of normal genes that promote excessive or inappropriate cell proliferation.

• Various RNA and DNA viruses have been associated with human malignancies (e.g., Epstein-Barr virus, certain papillomaviruses).

• Viruses carry viral oncogenes into target cells, where they become firmly established. Clonal descendants then carry the viral genes, which maintain the malignant phenotype of the cell clones.

• A very different class of cancer genes was discovered rather recently. Tumor-suppressing genes (antioncogenes) in normal cells appear to regulate the proliferation of cell growth. When this type of gene is inactivated, a block to proliferation is removed and cells begin a program of deregulated growth, or the genetically depleted cell itself may proliferate uncontrollably.

• No single satisfactory explanation exists for the success of tumors in escaping the immune rejection process. It is believed that early clones of neoplastic cells are eliminated by the immune response.

• Cells, rather than immunoglobulins, are believed to dominate tumor immunity.

• Four types of identified tumor antigens are tumor-specific antigens on chemically induced tumors, tumor-associated antigens on virally induced tumors, carcinofetal antigens, and spontaneous tumor antigens.

• A tumor marker is a characteristic of a neoplastic cell that can be detected in plasma or serum. Markers may be useful in the diagnosis and selection of different treatment approaches, monitoring therapies, and determining prognosis.

• Tumor markers include CEA, AFP, β-hCG, neuron-specific enolase, prostatic acid phosphatase, and placental alkaline phosphatase.

• Various modalities are used to treat cancer. In addition to the classic therapies, newer therapies (e.g., monoclonal antibodies) are being used.