The fundamental event that causes cells to become malignant is an alteration in their DNA structure which results in unrestrained cellular proliferation.

Most of these alterations involve sequence changes in DNA (i.e., mutations).

They may originate as a consequence of random replication errors, exposure to carcinogens (e.g., radiation), or faulty DNA repair processes. 1

Cancer is of clonal origin, with a single abnormal cell, often with multiple genetic alterations, multiplying to become a mass of cells forming a tumor. The tissue microenvironment influences the processes that occur.

The exact nature of these influences may vary with cell types involved, intercellular interactions and presence of factors such as paracrine signals, local hypoxia and proinflammatory responses. Carcinogenesis is, thus, a multistep process, ultimately transforming normal cells into malignant ones. Hence, tumors often take from a few to tens of years to develop to macroscopic levels.1 The transformation from a normal cell into a tumor cell is a multistage process, typically a progression from a precancerous lesion to malignant tumors.

The transformation of normal to neoplastic cells is caused by both endogenous and exogenous factors, including chemical and physical agents, viruses, activation of cancer-promoting genes, and inhibition of cancer-suppressing genes. 

Four classes of genes, can result in the development of a tumor.

These are proto-oncogenestumor suppressor genes, DNA repair genes, and genes regulating apoptosis or evasion of immune surveillance. When mutated to cause gain-, or loss-of-function or inappropriate regulation cause cancer.

The process is characterized by changes at the cellular, genetic, and epigenetic levels and abnormal cell division, in some cancers forming a malignant mass.

According to the prevailing accepted theory of carcinogenesis, the somatic mutation theory, mutations and epimutations in DNA that lead to cancer disrupt these orderly processes by disrupting the programming regulating the processes, upsetting the normal balance between proliferation and cell death. This results in uncontrolled cell division and the evolution of those cells by natural selection in the body. Only certain mutations lead to cancer whereas the majority of mutations do not.

 

Content 2

Content 3

 

-----------------

Variants of inherited genes may predispose individuals to cancer. In addition, environmental factors such as carcinogens and radiation cause mutations that may contribute to the development of cancer. Finally random mistakes in normal DNA replication may result in cancer causing mutations.[1] A series of several mutations to certain classes of genes is usually required before a normal cell will transform into a cancer cell.[2][3][4] On average, for example, 15 "driver mutations" and 60 "passenger" mutations are found in colon cancers.[2] Mutations in genes that regulate cell division, apoptosis (cell death), and DNA repair may result in uncontrolled cell proliferation and cancer.

Cancer is fundamentally a disease of regulation of tissue growth. In order for a normal cell to transform into a cancer cell, genes that regulate cell growth and differentiation must be altered.[5] Genetic and epigenetic changes can occur at many levels, from gain or loss of entire chromosomes, to a mutation affecting a single DNA nucleotide, or to silencing or activating a microRNA that controls expression of 100 to 500 genes.[6][7] There are two broad categories of genes that are affected by these changes. Oncogenes may be normal genes that are expressed at inappropriately high levels, or altered genes that have novel properties. In either case, expression of these genes promotes the malignant phenotype of cancer cells. Tumor suppressor genes are genes that inhibit cell division, survival, or other properties of cancer cells. Tumor suppressor genes are often disabled by cancer-promoting genetic changes. Finally Oncovirinaeviruses that contain an oncogene, are categorized as oncogenic because they trigger the growth of tumorous tissues in the host. This process is also referred to as viral transformation.

 

 

Proto-oncogenes

Proto-oncogenes are genes commonly used during normal growth and development; without control, they have the potential to produce neoplasms through their uncontrolled expression.

Oncogenes

The genes that can promote cell growth when altered are often called oncogenes. They were first identified as critical elements of viruses that cause animal tumors; it was subsequently found that the viral genes had normal counterparts with important functions in the cell and had been captured and mutated by viruses as they passed from host to host. Examples include chronic myeloid leukemia (abl), about half of melanomas (braf), Burkitt’s lymphoma (c-myc), and subsets of lung adenocarcinomas (egfr, alk, ros1, and ret).

Oncogenes are genes that have made the transition and are now capable of producing neoplasms. Most commonly, oncogenes cause unregulated cell growth through promotion of cellular division, which results in further mutations.

Tumor suppressor genes

Tumor suppressor genes are genes that function to help control cell growth; their loss thus results in uncontrolled cell growth through loss of regulation of division.

The p53 gene is the best-established tumor suppressor gene identified to date, and the normal wild-type gene appears to play an important role in suppressing neoplastic transformation. Of note, p53 is mutated in up to 50% of all human solid tumors, including liver, breast, colon, lung, cervix, bladder, prostate, and skin.

 

 

Apoptosis genes.

DNA repair genes.

 

Methods of conversion of proto-oncogene to oncogene

  • Overexpression of the gene.
  • Amplification of the gene.
  • Point mutation in the gene.
  • Translocation of the gene to another region with resultant overexpression of the gene, or resultant production of protein with oncogenic activity.

Role of oncogenes

Overview: Once converted from proto-oncogenes, oncogenes function by synthesizing growth factors, growth factor receptors, signal-transducing proteins, and nuclear transcription factors, or by promoting loss of regulation of cyclins and cyclin-dependent kinases.

  1. Synthesize growth factors to which the neoplastic cell is also responsive. For example, glioblastomas produce platelet-derived growth factor ( PDGF).

  2. Synthesize growth factor receptors. For example,

    • RET receptor for glial cell line-derived neurotrophic factor—in medullary and papillary thyroid carcinoma (MEN syndrome).
    • ERB B1, an epidermal growth factor (EGF) receptor, is overexpressed in squamous cell carcinoma of the lung.
    • ERB B2, an EGF receptor, is overexpressed in 25% of breast carcinomas.
  3. Synthesize signal-transducing proteins: An example of a specific gene is the RAS gene.

    • Incidence of mutations in RAS gene: Mutations of the RAS gene are in 30% of all malignant neoplasms and in 90% of pancreatic adenocarcinomas.
    • Role of normal RAS gene: The RAS gene codes for protein that is associated with a growth factor receptor. When stimulated by a growth factor, RAS binds guanosine triphosphate (GTP) and activates the mitogen-activated protein (MAP) kinase pathway, which results in activation of transcription. The RAS protein is controlled by its GTPase activity; it cleaves the GTP bound to it to guanosine diphosphate (GDP), which inactivates the RAS protein.
    • Effects of mutations in the RAS gene: The RAS protein loses its GTPase activity, so it remains activated, resulting in continual promotion of transcription.
  4. Synthesize nuclear transcription factors: An example of a specific gene is the MYC gene.

    • Neoplasms associated with mutations of the MYC gene: Burkitt lymphoma; also amplified in breast, lung, and colon cancers.
    • Role of normal MYC gene: MYC protein binds to DNA and activates transcription of several genes, including cyclin-dependent kinases (CDK). CDK proteins help drive the cell through the cell cycle.
    • Effect of activation to oncogene: Overexpression of MYC results in overpromotion of the cell cycle.
  5. Loss of regulation of cyclins and cyclin-dependent kinases.

Mutations in tumor suppressor genes

Important point: The two-hit hypothesis implies that with many hereditary neoplasms a tumor suppressor gene is involved. The protein product from one gene is enough to prevent neoplasms from developing; however, individuals born with a mutation of one gene are one step closer to the development of a neoplasm than those born with two normal genes.

Select tumor suppressor genes: Within neoplasms, the most common tumor suppressor genes with mutations are retinoblastoma and p53.

  1. Retinoblastoma (RB) gene

    • Associated neoplasms: Familial retinoblastoma and osteosarcoma; breast cancer and small cell lung carcinoma.
    • Role of normal RB gene: Retinoblastoma binds E2F transcription factor, which is needed for the cell to move from the G1 phase of the cell cycle to the S phase. When retinoblastoma is phosphorylated, the E2F is released and the cell moves through the cell cycle.
    • Effect of mutations of RBgene: Can affect retinoblastoma or the proteins that phosphorylate retinoblastoma, resulting in hyperphosphorylation of RB.
  2. p53 gene

    • Incidence: Mutations of the p53 gene are found in more than 70% of tumors.
    • Role of normal p53 gene
      • Activated by DNA damage.
      • p53 arrests the cell cycle by transcription of CDK1 (p21), which inhibits cyclin/CDK complexes and prevents phosphorylation of RB.
      • p53 promotes production of GADD45, which helps repair the cell.
      • If cellular damage is not repaired, p53 promotes induction of the Bax gene, which in turn promotes apoptosis.
  3. Other tumor suppressor genes include APC/β-catenin, INK4a/ARF, TGF-β, NF-1, VHL, and PTEN, as described below.

APC/β-catenin

  • Normal function of protein product: APC protein down-regulates β-catenin.
  • Effect of mutation: Elevated levels of β-catenin result in interaction with TCF, which results in increased levels of c-MYC and cyclin D1.
  • Incidence: Found in 70–80% of nonfamilial colon carcinomas; also found in 50% of hepatoblastomas and 20% of hepatocellular carcinomas.
++

INK4a/ARF

++
  • Normal function of protein product: Blocks cyclin D-CDK4 activity in the cell cycle.
  • Incidence: 20% of familial melanomas, 50% of pancreatic adenocarcinomas, and squamous cell carcinomas of the esophagus.

TGF-β (transforming growth factor beta)

++
  • Incidence: Inactivated in more than 70% of colon cancers in patients with hereditary nonpolyposis colon cancer (HNPCC) and in patients with sporadic colon cancer with microsatellite instability.
  • Associated mutation:SMAD4 (originally termed DPC4 [deleted in pancreatic cancer]). SMAD4 encodes part of the TGF-β growth inhibitory pathway. Mutations of SMAD4 are seen in 50% of cases of pancreatic adenocarcinomas.
++

NF-1

++
  • Normal function of protein product: Neurofibromin is a GTPase-activating enzyme.
  • Effect of mutation: RAS is trapped in an active form.
  • Associated neoplasms: Neurofibromas and malignant peripheral nerve sheath tumors.
++

VHL

  • Normal function of protein product: VHL protein is a ubiquitin ligase whose substrate includes HIF-1, which regulates vascular endothelial growth factor (VEGF).
  • Associated neoplasms: Nonfamilial renal cell carcinomas.
++

PTEN (phosphatase and tensin homologue)

++
  • Normal function of protein product: Blocks the cell cycle by increased transcription of p27.
  • Effect of mutation: Cells are allowed to easily progress into the cell cycle.
  • Incidence: Frequently found in endometrial carcinomas and glioblastomas; associated with Cowden syndrome.

Apoptosis genes

++
  1. Bcl-2

    • Normal function: Inhibitor of apoptosis.
    • Method of activation: Often a translocation of bcl-2 gene adjacent to a more heavily used gene, such as the immunoglobin ( Ig) heavy chain gene.
    • Consequence of activation: Increased production of bcl-2, resulting in inhibition of apoptosis.
    • Associated neoplasm: Follicular lymphoma.
  2. p53

    • Normal function: Promotes production of Bax (a pro-apoptotic gene).
    • Consequence of mutation: Less p53 results in less Bax, which indirectly causes inhibition of apoptosis.
++

DNA repair defects

++

Basic descriptions

++
  • Microsatellites: Repeats of 1–6 nucleotides in the genome; these are fixed and do not change.
  • Microsatellite instability: Changes in microsatellites, indicative of a DNA repair defect.

DNA repair defects occur in one of three systems: Recombination repair, mismatch repair, or nucleotide excision.

Development of ability to invade and metastasize

  1. Detachment of cells (example method: down regulation of e-cadherin seen in colon and breast carcinomas).

  2. Attachment to matrix (general method: expression of increased numbers of laminin and fibronectin receptors).

  3. Degradation of extracellular matrix (general method: production of metalloproteinases).

  4. Migration of tumor cells (general method: increased expression of CD44 adhesion molecule, which is used by T lymphocytes to migrate to lymph nodes).

 

For a cell to change from a normal cell to a malignant cell, it must:

  1. Acquire self-sufficiency in growth signals and ignore growth-inhibitory signals.

  2. Evade apoptosis, since apoptosis is the body's mechanism to rid itself of cells with genetic damage so they cannot propagate that damage.

  3. Acquire defects in DNA repair.

  4. Acquire the ability to divide an unlimited number of times.

  5. Promote angiogenesis.

  6. Invade surrounding tissue, passing through the basement membrane and spreading to distant organs (i.e., metastasize).

 

Cancer Stem Cells

 

  • Eradication of the neoplasm requires removal of the stem cells.
  • Stem cells must have the BMI1 gene—the protein product of the BMI1 gene inhibits p16INK4a and p14ARF. p16INK4a and p14ARF normally function to inhibit the cell cycle.

Cancers are named based on their origin: those derived from epithelial tissue are called carcinomas, those derived from mesenchymal tissues are sarcomas, and those derived from hematopoietic tissue are leukemias, lymphomas, and plasma cell dyscrasias (including multiple myeloma).

References

http://www.emedicinehealth.com/slideshow_pictures_cancer_101/article_em.htm#

All tumors have two basic components: (1) neoplastic cells that constitute the tumor parenchyma and (2) reactive stroma made up of connective tissue, blood vessels, and variable numbers of cells of the adaptive and innate immune system.

The classification of tumors and their biologic behavior are based primarily on the parenchymal component, but their growth and spread are critically dependent on their stroma. In some tumors, connective tissue is scant and so the neoplasm is soft and fleshy. In other cases, the parenchymal cells stimulate the formation of an abundant collagenous stroma, referred to as desmoplasia. Some demoplastic tumors—for example, some cancers of the female breast—are stony hard or scirrhous.

The terms benign and malignant correlate to the course of the neoplasm. Benign neoplasms stay localized in one place; malignant neoplasms invade surrounding tissue and, in most cases, can metastasize to distant organs. If the neoplasm is malignant, the cells must also gain the ability to invade the basement membrane and surrounding tissue, enter the blood stream, and spread to and grow within distant organs.

Monoclonal Origin

Cancers begin in a single stem cell and therefore are monoclonal in origin. A wide variety of genetic and epigenetic changes can occur in different cells within malignant tumors over time. Most cancers are characterized by marked heterogeneity in the populations of cells. The vast majority of human cancers are characterized by a multiple-step process involving many genetic abnormalities, each of which contributes to the loss of control of cell proliferation and differentiation and the acquisition of capabilities, such as tissue invasion, the ability to metastasize, and angiogenesis.

Immature Features

Cells that have undergone neoplastic transformation usually express cell surface antigens that may be of normal fetal type, and they may display other signs of apparent immaturity, and may exhibit qualitative or quantitative chromosomal abnormalities, including various translocations and the appearance of amplified gene sequences.

It is now well established that a small subpopulation of cells, referred to as tumor stem cells, reside within a tumor mass. They retain the ability to undergo repeated cycles of proliferation as well as to migrate to distant sites in the body to colonize various organs in the process called metastasis. Such tumor stem cells thus can express clonogenic (colony-forming) capability, and they are characterized by chromosome abnormalities reflecting their genetic instability, which leads to progressive selection of subclones that can survive more readily in the multicellular environment of the host. This genetic instability also allows them to become resistant to chemotherapy and radiotherapy. The invasive and metastatic processes as well as a series of metabolic abnormalities associated with the cancer result in tumor-related symptoms and eventual death of the patient unless the neoplasm can be eradicated with treatment.

Many cancers go through recognizable steps of progressively more abnormal phenotypes: hyperplasia, to adenoma, to dysplasia, to carcinoma in situ, to invasive cancer with the ability to metastasize. For most cancers, these changes occur over a prolonged period of time, usually many years.

Stem cells that initiate and sustain a neoplasm.

  • Eradication of the neoplasm requires removal of the stem cells.
  • Stem cells must have the BMI1 gene—the protein product of theBMI1 gene inhibits p16INK4a and p14ARF. p16INK4a and p14ARF normally function to inhibit the cell cycle.

This heterogeneity significantly complicates the treatment of most cancers because it is likely that there are subsets of cells that will be resistant to therapy and will therefore survive and proliferate even if the majority of cells are killed. These mutations become ancestors of the entire future tumor cell population. Evolution to more undifferentiated cells reflects high mutation rates. Cancers are characterized by unregulated cell division, avoidance of cell death, tissue invasion, and the ability to metastasize.

No one mutation will result in a malignant neoplasm; malignant neoplasms result from the survival of cells that have accumulated multiple mutations.

Conversion of one of the two allelic genes from a proto-oncogene to an oncogene is sufficient to promote neoplasia. However, it requires loss of both tumor suppressor genes to promote neoplasia, as one of the two genes is sufficient to produce enough product to inhibit neoplasia.

While most cancers arise sporadically, familial clustering of cancers occurs in certain families that carry a germline mutation in a cancer gene.Ibid

{Many new drugs either recently approved or in late stages of evaluation were designed to block the fundamental mutations that cause specific cancers: aberrant growth factor receptors, dysregulated intracellular signaling pathways, defective DNA repair and apoptosis, and tumor angiogenesis.

The primary tools for inhibiting these new targets are either monoclonal antibodies that attack cell surface receptors and antigens, or synthetic small molecules that enter cells and engage critical enzymes. }

 

 

 

 

Cancer develops as age advances

C

 

Cancer genetics is comprised of two main subfields: genetic susceptibility and somatic cell genetics.

Genetic susceptibility focuses on inherited (constitutional or germline) genetic variation in cancer susceptibility genes, and the effects of that inherited variation on an individual’s lifetime cancer risk.

In contrast, somatic cell genetics focuses on mutations that arise in an individual’s cells during their lifetime and the role that those mutations play during tumor initiation and progression.

Underlining the common biological basis of the substantial gene mutations involved in cancer initiation and progression, several biological characteristics consistent with tumor growth and metastasis were termed “hallmarks.” Those are six common traits that direct the transformation of normal cells to malignant cells. They include (1) growth stimulation, (2) evasion of growth suppressors, (3) resistance to apoptosis, (4) replicative immortality, (5) induction of angiogenesis, and (6) activation of invasion and metastasis. To these features, other key hallmarks were added: abnormal metabolic pathways and evasion of the immune system along with genome instability and tumor-promoting inflammation.

The best-known example of the mechanism involved in cancer is the TP53 tumor suppressor protein in which a mutation leads to inhibition of this gene activity and enables the survival of cancerous cells through resistance to programmed cell death.

 

 

Genes are involved in carcinogenesis by virtue of inherited traits that predispose to cancer (e.g., altered metabolism of potentially carcinogenic compounds), mutation of a normal gene into an oncogene that promotes the conversion of normal cells into cancer cells, or inactivation of a tumor suppressor gene, which triggers malignant transformation. A critical gene related to cancer in humans is the tumor suppressor p53. This gene is not only essential for cell viability, but critical for monitoring damage to DNA. Inactivation of p53 is an early step in the development of many types of cancer. Stimulation of oncogene formation by carcinogens (tobacco, alcohol, sunlight) is estimated to be responsible for 80% of cancers in the United States. Tobacco accounts for more cases of cancer than all other known carcinogens combined.

Certain heritable conditions predispose to the development of malignancy. These hereditary causes of neoplasms may be grouped into autosomal dominant neoplasia syndromes (Table 4-1) and defective DNA repair syndromes (Table 4-2), most of which are autosomal recessive. Importantly, although certain mutations are associated with the development of certain malignancies, one mutation alone is NOT enough to result in the development of a neoplasm.

Cancer cells must evade the host's immune surveillance system, which is designed to seek out and destroy tumor cells. Most mutant cancer cells stimulate the host's immune system to form antibodies. This protective role of the immune system is apparent in those with acquired immunodeficiency syndrome and recipients of organ transplants who are maintained on long-term immunosuppressive drugs. These groups have a higher incidence of cancer.

 

Hematopoietic stem-cell population dynamics may precede many hematologic cancers, including myeloproliferative neoplasms,3 myelodysplastic syndromes,4 acute myeloid leukemia (AML),5,6 and chronic lymphocytic leukemia.7 For example, in some patients, stem cells carrying a subset of the mutations present in the cancer cells are able to survive chemotherapy; subsequently, these cells acquire novel mutations, triggering a relapse.8-10 This suggests that a clonally expanded stem-cell population may have existed before the cancer developed.

Clonal mosaicism for large chromosomal abnormalities, reflecting expansion of a specific cellular clone, appears to arise in approximately 2% of elderly persons and is a risk factor for later hematopoietic cancers.11-15 In principle, clonal expansion among hematopoietic stem cells — a phenomenon termed “clonal hematopoiesis” — could be much more common16 and only occasionally accompanied by chromosomal abnormalities.

Many studies today sequence blood-derived DNA from thousands of persons to identify inherited risk factors for common diseases. We reasoned that such data offered the opportunity to test the hypothesis that clonal expansions with somatic mutations are common and often precede blood cancers and to identify the genes in which mutations drive clonal expansions.

 

It is generally believed—although not conclusively proved—that genetic alterations underlie all cellular and biochemical aberrations responsible for the malignant phenotype.

In addition to mutational changes that alter the genetic code, epigenetic changes also underlie cellular and biochemical aberrations that contribute to the malignant phenotype. Epigenetic phenomena influence gene expression and cell behavior, and although once acquired they are transmitted to daughter cells with cell division, they are not changes in the genetic code. An example of this is the silencing of certain genes by hypermethylation of DNA in the promoter region.

Although the progressive phenotypic characteristics of neoplasia result predominantly through sequential molecular alterations and abnormal function of the proliferating tumor cells, it is now clear that at some level abnormal function of the host stromal cells is fundamentally involved in continued tumor progression. It is not clear whether the abnormal function of stromal cells in tumor progression is due to genetic changes in these host cells or whether it is through cell-to-cell communications established through juxtacrine signaling loops with tumor cells. Stromal cell abnormalities can be nonproliferative, such as secretion of requisite growth factors, or proliferative, such as an expansion of the blood vessel network to support the growth of enlarging tumors.

Cells that have undergone neoplastic transformation usually express cell surface antigens that may be of normal fetal type, may display other signs of apparent immaturity, and may exhibit qualitative or quantitative chromosomal abnormalities, including various translocations and the appearance of amplified gene sequences. It is now well established that a small subpopulation of cells, referred to as tumor stem cells, reside within a tumor mass. They retain the ability to undergo repeated cycles of proliferation as well as to migrate to distant sites in the body to colonize various organs in the process called metastasis. Such tumor stem cells thus can express clonogenic (colony-forming) capability, and they are characterized by chromosome abnormalities reflecting their genetic instability, which leads to progressive selection of subclones that can survive more readily in the multicellular environment of the host. This genetic instability also allows them to become resistant to chemotherapy and radiotherapy. The invasive and metastatic processes as well as a series of metabolic abnormalities associated with the cancer result in tumor-related symptoms and eventual death of the patient unless the neoplasm can be eradicated with treatment.

Genetic Changes in Neoplasia

Maintaining genomic integrity is a fundamental cellular task. A complex cellular apparatus serves to recognize DNA damage or errors in replication, activate checkpoints to halt further cell replication, and implement corrective measures or signal suicidal cell death. One of the earliest phenomena observed in the course of tumor initiation is the development of defects in the genes involved in the machinery that guards the genome. This malfunction creates a degree of instability inherent in the genome that greatly increases the spontaneous rate at which genomic mutations or structural alterations occur and subsequently enables tumors to potentially acquire defects in an unlimited number of additional genes that may confer to them a growth advantage. Exposure to ionizing radiation and chemical carcinogens are environmental factors that can accelerate the accumulation of deleterious mutations. The cataloguing of these mutated genes has been a fundamental task of molecular oncology because it identifies genes whose functions are relevant to tumor cells. Genes that confer a growth advantage to tumor cells through a loss-of-function alteration are named tumor suppressor genes. Genes that confer a growth advantage through a gain-of-function event are named proto-oncogenes, and their activated counterparts are named oncogenes. Tumor suppressor genes can be inactivated through frame-shift mutation, deletion of part or all of the gene, and gene silencing by way of promoter methylation. Proto-oncogenes can be activated through mutation, gene amplification and overexpression, chromosomal translocation, and possibly other mechanisms.

Representative Oncogenes Activated in Human Tumors.

Oncogene Cellular Function Tumor Types Activated Mechanism of Activation
EGFR/HER1 Growth factor receptor Glioblastoma, lung and breast cancer Mutation, amplification
HER2/Neu Growth factor receptor Breast, ovarian, gastric cancer Amplification
PRAD1/Cyclin D1 Cell cycle regulator Breast and esophageal cancer, lymphoma, parathyroid adenoma Amplification, translocation
K-Ras, N-Ras, H-Ras G protein, signal transduction Multiple tumor types Mutation
B-Raf Signal transduction Multiple tumor types, melanomas Mutation
Src Adhesion and cytoskeletal signaling, other functions Colon, breast, lung cancer, sarcoma, melanoma Unknown, rarely mutated
Myc Transcription factor Multiple tumor types Amplification, mutation
Myb Transcription factor Leukemia Amplification, overexpression
Fos Transcription factor Multiple tumor types Overexpression
Int2/FGF3 Growth factor Esophageal, gastric, head and neck cancers Amplification
Fes/Fps Signal transduction Leukemia Unknown
menin Transcription factor Pituitary, pancreas , parathyroid tumors Mutation
Ret Growth factor receptor Parathyroid, medullary thyroid carcinoma, pheochromocytoma

Representative Tumor Suppressor Genes Inactivated in Human Tumors or the Human Germline.

Tumor Suppressor Gene Cellular Function Tumor Types Inactivated Mechanism of Inactivation Hereditary Syndromes with a Germline Inactivated Allele
p53 Cell cycle regulator Multiple tumor types Mutation Li-Fraumeni
Rb Cell cycle regulator Retinoblastoma, small cell lung cancer, sarcoma Deletion, mutation Familial retinoblastoma
APC Cell adhesion Colon cancer Deletion, mutation Familial adenomatous polyposis
PTEN Signal transduction, adhesion signaling Glioblastomas, prostate cancer, breast cancer Deletion, mutation Cowden’s
hMSH2 DNA mismatch repair Colon cancer, endometrial cancer, melanoma Mutation Hereditary nonpolyposis colon cancer
hMLH1 DNA mismatch repair Colon cancer, melanoma Mutation Hereditary nonpolyposis colon cancer
BRCA1 DNA ds-break repair Breast and ovarian cancers Mutation Familial breast/ovarian
BRCA2 DNA ds-break repair Breast and ovarian cancers Mutation Familial breast/ovarian
WT-1 Transcription factor Wilms’ tumor Deletion, mutation Childhood Wilms’ tumor
NF-1 GTPase activator Sarcoma, glioma Deletion, mutation Neurofibromatosis
NF-2 Cytoskeletal protein Schwannoma Mutation Neurofibromatosis
VHL Ubiquitin ligase Kidney cancer, multiple tumor types Mutation Von Hippel-Lindau disease
p16/CDKN2 Cell cycle regulator Melanoma, pancreatic and esophageal cancers

In general, during the gain-of-function alteration of a proto-oncogene, only one allele is mutated. In contrast, during the loss-of-function alteration of a tumor suppressor gene both alleles need to be inactivated. In certain cases, loss of one allele can result in reduction of gene expression. For some genes, this gene-dosage reduction is sufficient to permit tumorigenic growth.

In addition to being generated through the mutation of cellular proto-oncogenes, oncogenes can also be acquired through the introduction of foreign genomic material, typically transmitted by viruses. Although virally induced tumors are common in animals, only a few human tumors are directly caused by viral infection. Causative viruses and their associated malignancies are listed in Table 5–3. One such virus, human T-cell leukemia virus, is closely related to HIV and can cause a type of T-cell leukemia as a result of proteins encoded by the viral genome that are able to activate latent human genes. Human papillomavirus has long been linked epidemiologically to cervical cancer, and the serotypes most often linked have been found to encode proteins that can bind and inactivate host tumor suppressor gene products. In this situation, a causative gene is not necessarily introduced by the virus, but the viral genome is able to direct the inactivation of tumor suppressor gene products and thereby favor growth and proliferation as well as malignant potential. The ability of viruses to modulate the host cellular machinery—and in some cases retain altered mammalian genes that are oncogenic—is likely to have developed over the course of mammalian evolution, because an actively proliferating cell provides the optimal conditions for replication of virions and propagation of viral infections.

Oncogenic Human Viruses.
Virus Type Virus Family Associated Cancer Type
HTLV-I Retrovirus (RNA virus) T-cell leukemia/lymphoma
Hepatitis B Hepadnavirus (hepatotropic DNA virus) Hepatocellular carcinoma
Hepatitis C Hepadnavirus Hepatocellular carcinoma
Epstein-Barr Herpesvirus (DNA virus) Nasopharyngeal carcinoma
Burkitt’s lymphoma
Immunoblastic lymphoma
Hodgkin’s disease
HHV-8 (KSHV) Herpesvirus Kaposi’s sarcoma
Body cavity lymphoma
HPV serotypes 16, 18, 33, 39 Papillomavirus (DNA virus) Cervical carcinoma
Anal carcinoma
HPV serotypes 5, 8, 17 Papillomavirus Skin cancer

Key: HTLV, human T-cell leukemia-lymphoma virus; HHV-8, human herpesvirus-8; KSHV, Kaposi’s sarcoma herpesvirus.

The diploid human genome naturally contains defective alleles of many genes, and although defective alleles are for the most part biologically silent, in the case of tumor suppressor genes, a defective allele can confer significant cancer risk to an individual and all family members harboring such an allele. The loss of function of a gene in adult tissues is statistically much more probable when only one functional allele exists in all cells from the beginning of life, and inherited susceptibility to cancer is almost always a result of germline passage of a defective tumor suppressor gene allele. Many of the identified tumor suppressor genes that are frequently inactivated in sporadic human tumors have also been linked to specific hereditary cancer syndromes. In families with these syndromes, a defective allele of the responsible tumor suppressor gene is passed in the germline, and members who harbor this heterozygous  genotype inherit a high risk for tumors in which the second allele has also been lost. An inherited mutation in one allele of the p53 gene can cause the rare Li–Fraumeni syndrome, characterized by the early development of bone, breast, brain, and soft tissue tumors (sarcomas) along with other organ-specific tumors (such as adrenal cancer). Inherited mutations in single alleles of the BRCA1 or BRCA2 genes confer a high risk for breast or ovarian cancers. The hereditary cancer syndromes linked with many tumor suppressor genes are listed in Table 5–2. In contrast to single alleles of defective tumor suppressor genes, single alleles of mutationally activated oncogenes are not biologically silent and, if present in the germline, can have profound clinical manifestations, even embryonic demise. Because of this fact, inherited syndromes related to germline transmission of activated oncogenes are much more uncommon. A rare example, however, is the familial syndrome of multiple endocrine neoplasia type II, in which heterozygotes carrying an activated RET oncogene on chromosome 10 are at increased risk of developing two rare neural crest tumors: pheochromocytoma and medullary carcinoma of the thyroid, together with parathyroid tumors.

Proto-oncogenes & Tumor Suppressor Genes in Normal Physiology & Neoplasia

Proteins encoded by proto-oncogenes and tumor suppressor genes perform diverse cellular functions. Not surprisingly, these include proteins that recognize and repair DNA damage, proteins that regulate the cell cycle, proteins that mediate growth factor signal transduction pathways and that regulate programmed cell death, and proteins involved in cell adhesion, proteolytic proteins, and transcription factors. The function of many proto-oncogenes and tumor suppressor genes remains unknown. Mutations that confer selective advantage to tumors are those that result in increased genomic instability, elimination of cell cycle checkpoints, inactivation of programmed cell death (apoptotic) pathways, increased growth factor signaling, decreased cell adhesion, and increased extracellular proteolysis. The expression and functions of many genes can be simultaneously affected through deregulation of transcription factors. With rapid advances in sequencing technologies and high-throughput capabilities to study normal and tumor genomes, aggressive efforts are in progress to identify all the tumor suppressor genes and proto-oncogenes in the human genome.

Tumor suppressor genes include proteins involved in DNA damage control, cell cycle control, programmed cell death, and cell adhesion. Examples include both the retinoblastoma protein and the p16 cell cycle inhibitor, which function in regulation of the G1 checkpoint of the cell cycle. Loss of these genes can result in unchecked progression through the G1/S checkpoint. The p53 tumor suppressor gene is a critical guardian of genomic integrity and serves to recognize DNA damage and consequently inhibit cell cycle progression and induce programmed cell death. Loss of p53 can result in continued cell replication despite DNA damage and failure to activate programmed cell death. The fundamental importance of p53 function and of genomic stability in the oncogenic process is underscored by the fact that p53 mutations are the most common mutations in human cancers and are seen in more than half of all human tumors. The PTEN tumor suppressor gene is a phosphatase involved in the regulation of an important survival signaling pathway. Loss of PTEN function can result in unopposed survival signaling and failure to activate programmed cell death. Cadherins are proteins involved in cell–cell adhesion. Loss of cadherins can result in reduced cell adhesion, cell detachment, and metastasis. Table 5–2 presents a small list of examples of tumor suppressor genes. When fully identified, the entire list of human tumor suppressor genes will be much larger.

++

Proto-oncogenes include proteins involved in various steps of the extracellular growth factor signaling pathway from the membrane receptors to the membrane intermediates to the proteins mediating the cytoplasmic signaling cascades. The epidermal growth factor receptor (EGFR) binds a number of extracellular ligands and, in cooperation with its homolog, HER2, signals proliferative and apoptotic pathways. Overactivity of EGFR or HER2 can lead to unregulated control of growth and apoptotic signaling. The gene for EGFR or HER1 is mutated or amplified in nearly half of all glioblastomas, is amplified in a fraction of breast cancers and other epithelial cancers, and is mutationally activated in a fraction of lung cancers. The HER2 gene is amplified in 20% of breast cancers and confers a poorer prognosis. Ras is a membrane-bound signaling switch that functions immediately downstream of membrane receptors at a key branch point of cytoplasmic signaling. Mutational activation of Ras causes overactive cytoplasmic signaling and deregulation of proliferative and apoptotic pathways. Ras appears to be critically important in tumorigenesis because nearly one third of all human tumors harbor mutationally activated Ras. Raf is a serine-threonine kinase that functions downstream of Ras. Mutational activation of Raf similarly can lead to overactive signaling and deregulation of proliferative and apoptotic pathways and is commonly seen in many tumors. Table 5–1 presents a partial list of oncogenes identified in human malignancies, along with the tumor types in which they are commonly observed and the cellular function encoded by their proto-oncogene counterparts.

++

Another pathway frequently activated in many human cancers is the PI3 kinase signaling pathway. This pathway controls many cellular processes required for malignant transformation, particularly because it functions to allow the cell to deal with and respond to stress. Activation of this pathway allows cells to adapt to and survive in conditions of low oxygen, low nutrients, and other environmental stresses and signals processes leading to increased protein synthesis, increased energy production, use of alternative metabolic pathways, cell survival, and cell proliferation. This pathway can be activated by upstream signals or can be activated within the pathway by the mutational activation of PI3K or its downstream signal Akt or by mutational inactivation of its negative regulator PTEN.

++

It is now clear that the inactivation of a single tumor suppressor gene or the activation of a single oncogene is insufficient for the development of most types of human tumors. In fact, the process entails the sequential acquisition of a number of hits over a period of time leading to sequential cellular phenotypic changes from atypia to dysplasia to hyperplasia to in situ cancer to invasive and subsequently metastatic cancer. The largest body of evidence to support this theory has been generated from the molecular study of colon cancer and identifiable preneoplastic lesions, including adenomas and colonic polyps. In this model, the progressive development of neoplasia from premalignant to malignant to invasive lesions is associated with an increasing number of genetic abnormalities, including both oncogene activation and tumor suppressor gene inactivation. This theory is further supported by the identification of inherited abnormalities of several tumor suppressor genes, all associated with a strong familial tendency to develop colon cancer at a young age.

++

Some forms of human cancer appear to be more simplistic in evolution. A translocation of the long arm of chromosome 9 to the long arm of chromosome 22 leads to fusion of the BCR gene with the c-Abl gene and results in expression of the BCR-Abl oncoprotein seen in chronic myelogenous leukemia (CML). The expression of this oncogene in hematopoietic cells in animal models reproduces the disease. This oncogenic event is seen in virtually 100% of cases of this disease, and a treatment that inhibits the kinase activity of this oncoprotein produces remissions in nearly 100% of the patients. Thus, in contrast to the multistep process involved in most types of carcinogenesis, the steps necessary for the development of CML may be much simpler.

++

The identification of tumor suppressor genes and oncogenes as the fundamental enablers of tumorigenesis has led to the hypothesis that cancer can be successfully treated by treatments that counteract the biochemical sequelae of these molecular abnormalities. This has fueled attempts to develop therapeutic agents that can inhibit the function of activated oncoproteins or that can restore the function of inactivated tumor suppressor proteins.

++

Hormones, Growth Factors, & Other Cellular Genes in Neoplasia

++

Although structurally altered genes, classified as oncogenes or tumor suppressor genes, are key mediators of neoplasia, the role of unaltered genes is not to be dismissed and is likely equally important in carcinogenesis. Signaling proteins of all kinds may drive the oncogenic process through abnormal signaling: abnormal in time, duration, or intensity; abnormal tissue expression; or abnormal subcellular compartment localization. The regulation of growth in complex organisms requires specialized proteins for the normal growth, maturation, development, and function of cells and specialized tissue. The complexity of the human organism requires that these proteins be expressed at precisely coordinated points in space and time. An essential component of this regulation is the system of hormones, growth factors, and growth inhibitors. On binding to specific receptor proteins on the cell surface or in the cytoplasm, these factors lead to a complex set of signals that can result in a variety of cellular effects, including mitogenesis, growth inhibition, changes in cell cycle regulation, apoptosis, differentiation, and induction of a secondary set of genes. The actual end effects are dependent not only on the particular type of interacting factor and receptor but also on the cell type and milieu in which factor–receptor coupling occurs. This system allows for cell-to-cell interactions, whereby a factor secreted by one cell or tissue can enter the bloodstream and influence another set of distant cells (endocrine action) or act on adjacent cells (paracrine action). An autocrine action is also possible when a cell produces a factor that binds to a receptor on or in the same cell. Altered concentration of these growth factors as well as overexpression or mutations of the receptors can change the signaling behavior, contributing to a malignant phenotype. Only a subset of growth factor receptors are proto-oncogenes. However, many additional growth factors and growth factor receptors appear to be important in tumor growth and progression, although not classified as proto-oncogenes, because they serve tumorigenic causes without incurring mutations or without overexpression.

++

An important class of growth factor signaling molecules are the growth factor receptor tyrosine kinases (RTKs). A number of tyrosine kinase receptor families exist, and in experimental models most are capable of transforming cells if activated or overexpressed. Although all of these abnormalities are not necessarily seen in naturally occurring human tumors, these experimental data highlight the potential inherent in these proteins and the important role they may be playing in tumor cells despite lacking the oncogene label. Members of the HER family of RTKs are commonly mutated or amplified in human tumors and exemplify the important role of RTKs in human neoplasia. In many other tumors, they likely play an important role despite having a normal sequence and expression level. For example, HER1 (also called EGFR) is not mutated or overexpressed in colon cancers, but it is sometimes activated by autocrine signaling in the cancer cells, and EGFR-targeted therapies are used to treat this type of cancer. The platelet-derived growth factor (PDGF) receptors, fibroblast growth factor receptors, vascular endothelial growth factor receptors, and insulin-like growth factor receptor are all families of RTKs that function similar to HER family RTKs. These receptors are, in general, not reported to be mutated or amplified in human tumors. However, there is increased expression in many tumors or aberrant expression in tumors from tissue types that ordinarily would not be expected to express that receptor. Alternatively, excessive production of receptor ligands is due to a variety of mechanisms (ie, loss of epigenetic silencing of the gene coding for the ligand or excessive gene transcription of the same gene). In experimental systems, each of these RTK systems has oncogenic potential, building a circumstantial case that they may be important players in human tumors.

++

Some growth factor signaling pathways function to inhibit cell growth and provide negative regulation in response to extracellular stimuli. Desensitization of cells to such growth inhibitors is common in tumors. An example of this is the transforming growth factor-β (TGF-β). TGF-β has diverse biological effects. It potently inhibits cell proliferation but also stimulates the production and deposition of extracellular matrix (ECM) and adhesion factors. These functions are important in tissue remodeling during embryogenesis and wound repair. In some tumor types, the antiproliferative response to TGF-β is lost early on because of mutations in its downstream signaling components. However, continued secretion, and often oversecretion, of TGF-β by the tumor and stromal tissues leads to an increase in the production of ECM and adhesion factors and promotes the invasive and metastatic property of tumors.

++

Another important class of receptors is the large superfamily of nuclear hormone receptors. These include the cellular receptors for a variety of hormones, among them estrogen and progesterone, androgens, glucocorticoids, thyroid hormone, and retinoids. The actions of estrogen are fundamentally important in the development of breast cancer. In women, oophorectomy early in life offers substantial protection against the development of breast cancer, and in animal models mammary carcinogenesis is significantly retarded in the absence of estrogen. Approximately half of all breast cancers are dependent on estrogen for proliferation. Although these data clearly implicate the estrogen signaling pathway in breast carcinogenesis, specific abnormalities of the estrogen receptor (ER) are not seen in breast cancers; therefore, the ER does not qualify as a tumor suppressor protein or an oncoprotein. It is possible that, although the loss of certain tumor suppressor genes or activation of certain oncogenes leads to the development of breast cancer, continued ER function is essential throughout this process and without ER function it cannot proceed. Alternatively, it is possible that abnormal ER signaling, perhaps as a result of altered cofactors, cross-talk, or phosphorylation status, drives breast carcinogenesis. Although the mechanism by which estrogen and its receptor drive breast cancers has not yet been determined, its fundamental role in this disease is well established. Furthermore, treatments that work through inhibiting the production of the active ligand or that inhibit the function of the ER are the most effective therapies for breast cancer yet developed and are highly active in the prevention and treatment of breast cancer. The androgen receptor (AR), similarly, plays a critical role in the development of prostate cancer, although occasional activating mutations of the AR have been reported in prostate cancers. On the contrary, the ability of retinoids (ligands for retinoic acid receptors) that are well known to participate in the differentiation of a variety of tissues during development to cause the differentiation of certain tumors in tissue culture models has been exploited as a treatment approach for acute promyelocytic leukemia (APL). APL is characterized by a t(15;17) chromosomal translocation resulting in the fusion of the PML gene with the retinoic acid receptor-α (RAR-α) gene. The resulting fusion protein blocks the differentiation of hematopoietic progenitor cells and eventually leads to the development of APL. This fusion protein is not by itself transforming in experimental models and cannot be categorized as a classic oncogene or tumor suppressor gene, but it is etiologically involved in the pathogenesis of APL. Because the fusion protein contains the ligand-binding domain of RAR-α, it remains sensitive to ligand and treatment of patients with the ligand all-trans retinoic acid results in differentiation of tumor cells and complete remission in most patients with this disease.

++

Other functional membrane proteins not related to growth can also be present on tumors cells. The MDR-1 gene product belongs to a class of ATP-dependent channel transporter proteins and is present on some normal epithelial cells. Its physiologic role may be to pump toxic molecules out of the cell, but in some tumor cells, its overexpression causes efflux of certain chemotherapeutic agents, leading to drug resistance. In some situations, its expression can be induced by long-term exposure to chemotherapy.

Oncogenes and Proto-oncogenes

Molecular evidence suggesting the normal cellular origin of retroviral oncogenes was first obtained by showing that radiolabelled DNA from the avian retroviral oncogene src hybridized specifically to normal uninfected avian cellular DNA as well as to normal mammalian DNA and even normal human cellular DNA. All retroviral oncogenes are now known to have hybridizing homologs, that is, close relatives, in the genomes of virtually all normal vertebrate cells. The normal host cell homologs of v-onc genes are called c-onc genes or proto-oncogenes.

The retroviral homologs found in normal cellular DNA have the usual structure of cellular genes, possessing both exons and introns, whereas the oncogenes in retroviruses do not have introns. It has been concluded that the oncogene analogs found in normal cellular DNA represent native cellular DNA and are not of viral origin. It appears, then, that retroviral oncogenes are copies (allowing for subtle differences in gene sequences) of normal cellular genes which were picked up and transduced into the retroviral genome by pre-existing retroviruses. Nevertheless, retroviral oncogenes usually show structural mutations or changes in expression relative to the corresponding proto-oncogenes. Since proto-oncogenes are highly conserved in vertebrates and are demonstrable in the cellular genomes of virtually all metazoans, their roles in cellular functions are apparently of fundamental importance. The gene products of proto-oncogenes have been identified as proteins (growth factors, growth-factor receptors, signal transducer proteins) with known functions in normal cells.

Direct evidence for the activation of proto-oncogenes to transforming genes in-vivo, independently of any retroviral gene participation, has been obtained by isolating proto-oncogenes, attaching them to promotor or enhancer sequences, and introducing this DNA into one-cell mouse embryos (fertilized eggs) and thus ultimately the germ cells of so-called transgenic mice. The promotor causes a high rate of transcription ("activation") of the introduced proto-oncogene resulting in a high incidence of malignant tumors in some of the progeny mice. In other experiments, "weak" retroviruses which lack a separate viral oncogene have been shown to produce tumors only when, by chance, the proviral DNA is inserted into the cellular genome next to a cellular proto-oncogene, which is then activated through the effect of the inserted viral promoter.

In summary, the oncogene theory postulates that the oncogenes of transforming retroviruses are derived from normal cellular genes (proto-oncogenes); and that an increased expression (activation) of proto-oncogenes or an inappropriate expression of mutated forms of proto-oncogenes, occurring spontaneously or induced by cancer-causing agents, contributes to neoplastic transformation and the development of cancer, including human cancer.

It is now recognized, as you shall learn, that sequential mutations or inappropriate expressions of several different classes of cellular genes [oncogenes, tumor suppressor genes, DNA nucleotide mismatch repair genes, and genes that mediate programmed cell death (apoptosis)] are probably involved in the usual multiple-step process that leads to human cancer.

Oncogenes and Human Cancers


Most of the known retroviral oncogenes have hybridizing homologs in normal human cellular DNA (proto-oncogenes). Oncogenes are activated in somatic cells in many forms of human cancer, including carcinoma, sarcoma, leukemia, and lymphoma. The chief mechanisms of oncogene activation are chromosomal translocation, point mutation, and gene amplification (Table).

Table: Activation of Cellular Proto-oncogenes in Human Cancers

 

Proto-oncogene Activation by Chromosomal Change Associated Cancer

c-myc Genetic rearrangement Translocation: 8-14, 8-2, or 8-22 Burkitt's lymphoma
c-abl Genetic rearrangement Translocation: 9-22 Chronic myeloid leukemia
c-H-ras Point mutation Bladder carcinoma
c-K-ras Point mutation Lung and colon carcinoma
N-myc Gene amplification Neuroblastoma

    Burkitt's Lymphoma

    In the cells of this human B cell lymphoma there is often a translocation between chromosomes 8 and 14, or between 8 and 2 or 22. This change transposes the cellular myc gene, which is normally on chromosome 8, into proximity with a predictably active promotor (in B lymphocytes) involved in immunoglobulin (Ig) synthesis. (Ig heavy chain loci are on chromosome 14, Ig light chain loci are on chromosomes 2 and 22). The translocation interrupts the normal transcriptional control of the myc gene and leads to amplification of the amount of the myc-encoded protein, thereby thrusting the myc proto-oncogene into the role of an oncogene.

    Chronic Myelogenous Leukemia (CML)

    The leukemia cells of virtually all (90-95%) patients with CML have a characteristic cytogenitic abnormality called Philadelphia chromosome, Ph', (Nowell, P.C., and Humperford, D.A., Science 132:1497, 1960) resulting from a reciprocal translocation between the long arms of chromosomes 9 and 22 (Rowley, J.D., Nature 243:290, 1973).

    In this translocation, the proto-oncogene c-abl is translocated from its normal location on chromosome 9 to the break point region in chromosome 22, resulting in the generation of a fusion gene and its protein product (BCR-ABL), a constitutively activated mutant tyrosine kinase with proven oncogenic activity. Clinical trials with a selective inhibitor (imatinib, Gleevec) of BCR-ABL tyrosine kinase in CML provide cytogenetic and hematologic evidence of improvement in patients with the chronic phase of CML (Drucker, B.J., et al., N Eng J Med 344: 1031-37, 2001; Kantarjian, H., et al., ibid. 346:645-52, 2002).

    Human Carcinomas

    The cellular DNA of tumor cells can be extracted and, following co-precipitation with calcium phosphate, introduced into the nuclei of selected cells (usually the mouse fibroblast cell line NIH 3T3) in vitro, a process termed transfection. Those cells which take up intact segments of DNA that induced the neoplastic state become transformed themselves. One can then isolate and sequence the transforming genes from these cells. In one of the earliest reported studies, such a transforming gene was identified in cells of the human bladder carcinoma cell line T24. When sequenced, this human oncogene in T24 cells was found to be a homolog of the v-H-ras gene (known to induce sarcomas and leukemias in susceptible murine animals). Furthermore, the human oncogene differed from the corresponding normal human proto-oncogene by only a single base substitution (G for T), a point mutation leading to the substitution of a single aminoacid (valine for glycine) at position 12 of the 21,000 m.wt. encoded protein.

    Other transfection studies have shown that the transforming genes of a number of other human cancers are also homologs of members of the ras family: for example, the transforming genes of cell lines and primary carcinomas of human lung and colon are homologs of the v-K-ras gene. (as noted in the previous table)

    An important finding is that when primary cultures of mouse embryo fibroblasts, rather than established cell lines such as NIH 3T3, are used as the DNA recipients in transfection assays, at least two co-operating oncogenes (often myc and ras) are usually required for complete neoplastic or "tumorigenic" conversion. This observation is consistent with the prevailing view that carcinogenesis is a multiple-step process. At this writing, more than 60 different oncogenes have been characterized in avian and mammalian, including human, species.

    Role of Individual Cellular Oncogenes in Neoplastic Transformation

    One of the simplest classifications of oncogenes, on the basis of the localization of their encoded proteins either in the cytoplasm or the nucleus, is useful in grouping those with similar roles. The accompanying table indicates the cellular localization of proteins encoded by some cellular proto-oncogenes and, for comparison, transforming proteins encoded by oncogenic DNA viruses.

    Table: Proteins Encoded by Proto-oncogenes and DNA Tumor Viruses

     

    Localization Proteins Encoded by
    Cellular Proto-oncogene DNA Tumor Virus

    Nuclear myc SV40 large T
    N-myc Polyoma large T
    myb Adenovirus E1a
    Cytoplasmic ras Polyoma middle T
    abl
    src
    erb B
    sis

    Most of the nuclear cellular oncogenes, that is, those with nuclear localization of the encoded proteins, appear to have similar, although not identical, roles. The most predictably observed trait which, as oncogenes, they confer on the affected cells is "immortalization", or the ability to be passaged in cell culture continuously without limit. Related traits which appear to be conferred are an incresed plating efficiency and the ability of cells to divide in the presence of low concentrations (0.5%) of serum. Nuclear cellular oncogenes show little ability to confer anchorage independence (growth in the absence of an added support structure) on cells in which they are activated and likewise only minimally induce morphologic changes, such as multilayering, of cultured cells.

    Cellular oncogenes with cytoplasmic protein products are relatively "weak" in inducing immortalization of cells in culture, but, in contrast to nuclear oncogenes, are strong in conferring the morphologic changes of transformation as well as in conferring anchorage independence.

    Neither nuclear nor cytoplasmic cellular oncogenes, acting alone, appear able to induce full transformation of cells. Their actions in transformation appear to be complementary. Many types of cells have been transformed, and rendered "tumorigenic" in animals, by transfection with collaborating pairs (one nuclear, one cytoplasmic) of cellular oncogenes in various combinations.

    Conversion of Cellular Proto-oncogenes to Oncogenes

    The known mechanisms of proto-oncogene activation and well-studied examples include:

    • translocation: c-myc, c-abl;
    • promotor insertion: c-myc, c-erb B;
    • point mutation: c-ras;
    • deletional mutation: c-erb B;
    • amplification: c-myc.

    Nuclear oncogenes of cellular origin are converted from proto-oncogenes by processes (translocation, promotor insertion, amplification) which lead to an increased level (or amount) of their encoded proteins. The disordered regulation of c-myc expression related to the 8-14 chromosomal translocation in Burkitt's lymphoma is the best documented example. In this genetic rearrangement, the normal promotor-enhancer regulators for c-myc are removed and replaced by those from predictably active immunoglobulin genes, resulting in amplification of the amount of the myc-encoded protein.

    Most of the cytoplasmic oncogenes of cellular origin are derived from proto-oncogenes by mutations which alter the structure of the encoded proteins. The c-H-ras gene whose activation is associated with several human carcinomas is an important example. Point mutations of the proto-oncogene involving aminoacids at residues 12, 13, or 61 induce transformation even when the 21,000 m.w. protein, p21ras, is present at low levels. Further studies of cytoplasmic cellular oncogenes, such as src, erb B, and neu (related to erb B), show that structurally altered protein products cause transformation, while over-expression of the normal protein brings about little change.

    An important cytoplasmic proto-oncogene is the abl gene which is present at the site of the 9-22 translocation resulting in the formation of the Philadelphia (Ph') chromosome typically seen in chronic myelogenous leukemia (CML). The abl protein is present in increased amounts in the tumor cells, but, apparently more important, the protein is altered (the translocation removes its amino terminus). Over-expression of the normal intact abl protein seems to cause no effect.

    In summary, many cytoplasmic proto-oncogenes appear to be converted to oncogenes by mutations which alter the structure of their encoded proteins. In these circumstances, the amount of the normal protein appears to be of little importance in cellular transformation.

    While the genetic changes in Burkitt's lymphoma and chronic myelogenous leukemia occur in cells of hematologic origin, no single genetic abnormality has thus far been identified as the transforming event in common human cancers of epithelial origin.

    Cellular Functions of Oncogene-Encoded Proteins

    The functions of proteins encoded by selected protooncogenes (and oncogenes) are given in the accompanying table.

    Table: Cellular Functions of Proto-oncogene Encoded Proteins

     

    Function Protein Protoonco-
    gene
    Associated Human Cancers

    Growth factor PDGF sis Osteosarcoma
    Growth-factor receptor EGF receptor erb B Breast, lung, ovarian cancer
    Post-receptor signal transduction GTP-binding protein ras Lung, colon, pancreatic cancer
    Nuclear transcription regulator myc protein myc Breast, colon, lung cancer, Burkitt's lymphoma

    Most, if not all, of the many proteins encoded by cellular oncogenes are involved in the "growth factor-receptor-response" pathways of transmission of growth stimulatory signals from the cell surface to the nucleus, culminating in the transcription of certain genes and in DNA synthesis.

    Aberrations in the normal process of transmembrane signaling involving growth factors (GFs), GF receptors, post-receptor "transducer" proteins, and nuclear controls may be caused by activated oncogenes and result in the abnormal growth characteristics of neoplastic cells.

    • Growth factors (GFs) are ubiquitous polypeptides that are produced and secreted by cells locally and that stimulate cell proliferation by binding to specific cell-surface receptors on the same cells (autocrine or autostimulation) or on neighboring cells (paracrine stimulation).
    • Conceptually, uncontrolled autostimulation from the persistent over-production of GFs may convert a cell to the neoplastic state. Human platelet-derived growth factor (PDGF), so named because it is released from platelets during blood clotting, is a major growth factor recoverable from serum. PDGF is apparently encoded by c-sis, the normal analog of v-sis which is the transforming gene of simian sarcoma virus (SSV). Cultured cells infected with SSV produce a PDGF-like mitogen which binds to PDGF receptors and stimulates the cells to proliferate in an uncontrolled manner, resulting in transformation.
    • Structural changes or amplification of GF receptors may promote neoplastic transformation. If GF receptors are changed in a way that continually presents cells with growth stimulatory signals, even in the absence of GFs, cells may respond as though high levels of GFs were present. One of several examples, all in the group of cytoplasmic oncogenes, can be mentioned. A portion of the human epidermal, growth factor receptor protein (now called Her-2/neu or Her-2) is similar to the proto-oncogene encoded erb B protein. Her-2 protein is over expressed in tissue and serum of ~25-30% of patients with breast cancer. Trastuzumab (Herceptin) is a recombinant humanized monoclonal anitbody that selectively binds to and mediates antibody-dependent cytolytic destruction of Her-2 protein and is approved for use in patients with metastatic breast cancer whose tumors over express Her-2.
    • Autonomy to GFs may be produced by changes in post-receptor "transducer" proteins that enable them to transmit growth stimulatory signals without prompting by a GF receptor. Ras-encoded proteins are guanosine triphosphate (GTP)-binding proteins that function as post-receptor signal transducers (cyclic on-off switches) for growth stimulatory signals.. Ras proteins are located in the plasma membrane and mediate the passage of growth signals from outside to inside the cell and to the cell nucleus, thus initiatiating the cell cycle and DNA synthesis. Mutations in ras proteins, potentially associated with a continuous growth signal that cannot be deactivated, are found in about 30% of human cancers (Wittinghofer, F., Nature 394: 317, 1998).
    • The cell division cycle is normally monitored at critical check points along the mitogenic signaling pathway. Deregulation of the cell cycle at the critical transition phase from G1 (resting) to S (synthesis of DNA) occurs in many types of human cancer. Transcriptional amplification of myc and related nuclear proteins is also associated with unregulated cell proliferation and transformation. Additionally, the microtubule network is necessary for interphase and mitotic cellular events, and paclitaxel (Taxol) which disrupts microtuble assembly is used for adjuvant therapy in advanced cancers of breast, ovary and lung.

    In summary, the activation of proto-oncogenes to cancer-associated oncogenes represents a "gain of gene function" (dominant allele) mutation involving pathways of transmission and transduction of mitogenic signals from cell to cell and from cell surface to nucleus and the activation of certain nuclear genes, culminating in DNA synthesis, unregulated cell division, and neoplastic transformation.

Tumor Suppressor Genes


Most studies of genes and growth factors involved in carcinogenesis have focused on functions that have a positive stimulatory effect on cell proliferation and, hence, neoplastic transformation. While the activation of some genes may result in cancer, it is reasonable to suppose that the activation of other genes may suppress the development of cancer. The existence of tumor suppressor genes, or so-called "anti-oncogenes", in normal cells is suggested by the results of somatic cell hybridization experiments in which fusions of malignant tumor cells, including those with activated oncogenes, with normal diploid fibroblasts were usually found to yield non-malignant hybrid cells. It is possible, therefore, that the loss or inactivation of putative tumor suppressing genes may remove a block to cell proliferation and the development of the neoplastic state. Supporting evidence linking the loss of genes to the development of human cancer comes from studies of heritable cancers, among them retinoblastoma of childhood.

Retinoblastoma


Retinoblastoma, the most common primary malignant eye tumor of children, occurs in both a heritable (autosomally dominant) form characterized by early onset and bilateral eye involvement and a sporadic form with later onset and unilateral involvement.

Sequential deletions or mutational losses of function of both allelic genes ("two-hit" model) at the Rb locus on chromosome 13q14 are required for the development of retinoblastoma of either form. In this genetic model for the two forms of retinoblastoma, first proposed by Knudson and now a paradigm for some other forms of hereditary cancer, the first mutation (first "hit") affecting retinal cells may be one that is inherited at birth through the germ line (heritable form) or that is acquired later by somatic mutation (sporadic form). The second mutation (second "hit") at the same locus is somatic and random in either form, and both allelic (homozygous) mutations are necessary for neoplastic transformation of a retinal cell to retinoblastoma. The sequential mutations necessary for transformation of a retinal cell are much (>1000-fold) more likely to occur in a cell that already has one inherited mutant allele than in a cell that has none, consistent with the clinical characteristics of this and some other forms of hereditary cancer: early onset, multiple primaries, bilateral involvement of paired organs, and Mendelian pattern of inheritance.

Germ line inheritance of a mutated Rb allele also predisposes to the development of a second form of childhood cancer, notably osteosarcoma in adolescence. Somatic mutations (deletions, frameshifts, etc.) of Rb are also found in various forms of adult cancer of lung, breast, prostate, and other sites.

The normal (wild-type) Rb gene on human chromosome 13q14 is a transcription regulator, and its protein product pRb regulates a critical check point in the cell cycle, namely progression from a G1 (resting) phase to the S (DNA-synthetic) phase. Loss of function mutation of both Rb alleles removes this regulatory control.

There are many regulator controls in the cell cycle. Very briefly, cyclin proteins regulate timing mechanisms through CDKs (cyclin-dependent kinases) that enzymatically phosphorylate and activate proteins critical to the cell cycle, particularly at the transition phase from G1 (rest) to S (synthesis of DNA). pRb normally binds to and inactivates at least two protein enzymes that are essential for the synthesis of DNA precursors and the promotion of DNA replication. The hyper-phosphorylation of pRb by CDKs releases and reactivates these enzymes and thus abolishes the inhibitory control of pRb in the cell cycle.

The majority of human cancers appear to have alterations in at least one of the main regulatory controls of the G1-S transition point in the cell cycle (Russo, A.A., et al., Nature 395:237-43, 1998).

Colorectal Cancer


Carcinoma of colon and rectum ranks second to lung cancer as a cause of cancer mortality in males and third (after breast and lung cancer) in females and accounts for about 60,000 deaths annually in the U.S. The majority of colorectal carcinomas arise from adenomatous polyps (adenomas) that develop in the bowel mucosa.

Familial adenomatous polyposis (FAP), an autosomal dominant trait with a prevalence of about 1 in 10,000, predisposes to the development of multiple intestinal polyps and colorectal carcinoma with early age of onset (age 30 or so). Recent molecular genetic studies of FAP and its progression into cancer indicate that normal colonic epithelial cells evolve into polyps and subsequently into cancer through a sequence of mutations and interactions of two kinds of genes: tumor suppressor genes and oncogenes.

The first genetic change in the development of FAP is the mutational inactivation of a tumor suppressor gene called APC (adenomatous polyposis coli) on chromosome 5(q21) of normal colonic epithelial cells (Kinzler, K.W., et al. Identification of FAP locus genes from chromosome 5q21. Science 253:661-664, 1991). The subsequent progression of polyp into colonic cancer involves stepwise changes, beginning with mutational activation of the ras gene on chromosome 12 and followed by deletions of tumor suppressor genes on chromosomes 18 and17, and perhaps others. The observed sequence of mutations involving APC, ras, DCC ("Deleted in Colon Cancer"), and p53 is also found during tumor development in some, but not all, patients with non-familial (sporadic) colorectal cancer, indicating that this is not the only genetic pathway to this common cancer.

p53 Tumor Suppressor Gene


A normal (wild-type) nuclear protein of 53 kilodaltons called p53 protein is the product of the p53 tumor suppressor gene on human chromosome 17(p13). As previously noted with the Rb gene, mutated or deleted tumor suppressor genes such as mutant p53, while heritable as a dominant allele, are recessive to the normal (wild-type) allele in somatic cells, and mutations or deletions in both p53 alleles ("two hits") are required for loss of function. Patients with germline mutations at the p53 locus are at very high risk for cancer development, as seen in the Li-Fraumeni hereditary cancer syndrome characterized by early onset of breast carcinoma, childhood sarcomas, and other tumors.

Somatic mutations at the p53 locus, usually point mutations substituting one amino acid for another and inactivating suppressor activity, are the most common genetic change in human cancers and occur in about 50% of them, including carcinoma of breast, colon, stomach, bladder, and testis, melanoma, and soft-part sarcoma.

Normal p53 protein is a transcription factor (increases gene expression) and mediates several cellular functions: regulation of the cell division cycle, DNA repair, and programmed cell death. In response to various forms of genomic DNA damage (caused by oncogene activation, radiation, cytotoxic drugs, hypoxia, certain viruses), the p53 protein can arrest the cell cycle at the G1 to S transition point, thus affording time for DNA repair and preventing duplication of a mutant cell or, alternatively, failing DNA repair, p53 protein can implement programmed cell death (apoptosis). Accordingly, p53 has been dubbed the "guardian of the genome." The cellular process of DNA replication is initiated by the formation of complexes between proteins called cyclins and enzymes termed cyclin-dependent kinases (CDKs). The formation of cyclin-CDK complexes is inhibited by p53 protein and other cell-growth inhibitors.

As previuosly noted, mutated or deleted tumor suppressor genes, such as mutant p53 or mutant Rb, while heritable as a dominant allele are recessive to the normal (wild-type) allele in somatic cells, and mutations or deletions in both alleles are required for loss of gene function.

DNA Mismatch-Repair Genes


A new class of cancer susceptibility genes was recently identified in hereditary non-polyposis colorectal cancer (HNPCC):a defective hMSH2 gene located on human chromosome 2p and a defective hMLH1 gene located on chromosome 3p. These defective genes are associated with widespread instability of microsatellite DNA and other short repeat sequences in HNPCC cells and with the accumulation of mutations, both germ line and somatic, throughout the genome of some HNPCC patients (Peltomaki,P.,et al., Science 260: 810-812, 1993; Ionov,Y.,et al.,Nature 363:558-561, 1993).Other studies indicate that natural (wild-type) hMSH2 and hMLH1 are human homologs, respectively, of a bacterial gene and a yeast gene that encode a base-binding enzyme involved in DNA mismatch repair.Genetic defects in DNA repair genes not only contribute to the development of HNPCC but may also be involved in other hereditary cancer syndromes as well as in the genetic instability (heterogeneity) commonly shown by cancer cells.

It is estimated that about 0.5% of the general population (~ 1 million Americans) may carry this or other mutator genes, along with a greatly increased risk of colorectal carcinoma or other cancers, such as, ovarian, uterine, and renal.

Apoptosis


Apoptosis (Gr. apo, away; ptosis, falling) is a normal process of non-random cell death that occurs in many biological conditions, among them: embryological development; perinatal thymus selection and deletion of self-responsive T-cells; normal cell turnover throughout life; and in response to abnormal stimuli, such as genomic DNA damage, but without concurrent pathological necrosis and inflammation.

Apoptosis (programmed cell death) is an active, energy-dependent process characterized by the rapid occurrence of distinctive morphological and biochemical changes in the cell. These changes include: the formation of cytoplasmic blebs; chromatin condensation at the nuclear membrane; and cleavage of chromatin by an endonuclease that is exclusively activated in apoptosis and that yields a distinctive 'chromatin ladder' pattern of DNA fragments, as shown by gel electrophoresis. Specialized proteases called caspases (cysteine aspartases), associated with mitochondria and the cytochrome C respiratory pathway, accelerate the cell-death response (Earnshaw, W. C., Nature 397: 387-389, 1999).

As noted elsewhere (see: p53 Tumor suppressor gene), p53 protein expression in response to genomic DNA damage can arrest the cell cycle (at G1) for DNA repair, thus preventing duplication of a mutant cell or, alternatively, failing DNA repair, implement cell suicide through programmed cell death.

If oncogenes are likened to an "accelerator" of cell proliferation or transformation and tumor suppressor genes to a "brake", then apoptosis is a final "suicidal crash". Thus, at least two signals ( and obviously multiple genetic controls) are required for cell proliferation or transformation: one that drives cell proliferation; and one that blocks cell death. Surprisingly, the protein product of the myc proto-oncogene has one domain that mediates cell proliferation and another one that, in the absence of required growth factors, nutrients, or other gene products, induces apoptosis. On the other hand, the bcl-2 proto-oncogene (known to be activated by chromosomal translocation in a variety of B-cell lymphomas) encodes an antiapoptosis protein; the bcl-2 protein product functions as an inhibitor of apoptotic cell death.

Human Cancer Cells Created with Defined Genetic Elements


Sequential mutations or inappropriate expressions of different classes of cellular genes (oncogenes, tumor suppressor genes, mismatch repair genes, and genes that mediate apoptosis) are involved in the usually multiple-step process that leads to human cancer.

This concept was put to critical test in a recent landmark study: normal human epithelial cells (and fibroblasts) in culture were transformed into cancer cells by the insertion of defined genetic elements (Hahn, W.C., Counter, C. M., et al., Nature 400: 464-468, 1999). The three necessary genetic elements were serially inserted and included: a subunit of the telomerase gene, to immortalize the cells by telomere maintenance, i.e., preventing telomere shortening at each cell division (see: Neoplasia VIII. Chromosomal Abnormalities: Telomeres)); an activated ras oncogene, known to be mutated in many kinds of human cancers; and a viral oncoprotein gene (SV 40 large T), known to inhibit the tumor suppressor proteins p53 and pRb.

When injected into immunodeficient mice, the transformed cells formed malignant tumors having similar identifying cellular and molecular genetic characteristics as the injected cells, thus suggesting that the tumorigenic growth was not the consequence of some other rare random event occurring in vivo after inoculation of these cells. Telomere maintenance appeared to be essential for the formation of human cancer cells.

Cancer Susceptibility Genes - Summary


The following table gives only a short list of the ever increasing number of cellular genes that, through inherited or somatic mutations and a gain (dominant allele) or loss (recessive allele) of function, are associated with the development of certain forms of human cancer. Prototype examples of affected genes and associated cancers were previously discussed.

Activating (gain of function) somatic mutations of proto-oncogenes (such as ras, myc, abl, etc.,) are present in a variety of sporadic human cancers (Table). Surprisingly, inherited mutations of proto-oncogenes are not regularly found in hereditary cancers with the following notable exception: inherited germ line mutation in the ret (RET) proto-oncogene confers a genetic predisposition to multiple endocrine neoplasia.

Germ line and subsequently somatic (loss of function) mutations associated with human cancer susceptibility and expression (see Table) mainly involve tumor suppressor genes (established examples include: APC, Rb,& p53 which is mutated in many forms of cancer; BRCA1, BRCA2, NF1, WT1, and DNA repair genes (such as hMSH2).In familial breast cancer, an inherited germ line mutation of the BRCA1 gene located on chromosome 17(q21) confers a predisposition to early-onset (~ premenopausal) breast cancer and ovarian cancer; and an inherited mutation of the BRCA2 gene located on chromosome 13(q12-13 is also linked to early-onset breast cancer. Mutations in BRCA1 and BRCA2 are each estimated to account for less than 5% of the total of all female breast cancer cases (~180,000) occurring annually in the U.S.

Table: Cellular Genes Associated with Human Cancer Susceptibility and Expression

Human Genes Associated with Cancer Susceptibility and Expressions

Affected Gene Chromosome Associated Cancer

Oncogenes:
abl 9(q24) Chronic myeloid leukemia
c-myc 8(q24) Burkitt's lymphoma
ras 12(p) Variety of cancers: colon, lung, pancreas, leukemia
N-myc 2(p) Neuroblastoma, small cell cancer of lunh
RET 10(q11) Medullary thyroid carcinoma, multiple endocrine neoplasias
PML/RAR-alpha t(15;17) Acute promyelocytic leukemia
Tumor Suppressor Genes:
APC 5(q21) Colon carcinoma
BRCA 1 17(q21) Breast and ovarian carcinoma
BRCA 2 13(q12-13) Breast carcinoma
p53 17(p13) Variety of cancers, Li-Fraumeni syndrome
NF1 17(q11) Neurofibromatosis type 1
RB 13(q14) Retinoblastoma, osteosarcoma
WT1 11(p13) Wilms' tumor
Mismatch Repair Genes:
hMSH2 2(p16) Colon carcinoma

Cancer is a genetic disorder in which the normal control of cell growth is lost. At the molecular level, cancer is caused by mutation(s) in DNA, which result in aberrant cell proliferation. Most of these mutations are acquired and occur in somatic cells.

However, some people inherit mutation(s) in the germline.The mutation(s) occur in two classes of cellular genes:

oncogenes

tumor suppressor genes.

Transformation of proto-oncogene to oncogene is the result of gain in function through:

  • Over-expression of the gene, or duplication (such as amplification) to produce increased onco-protein
  • Activation or formation of fusion gene by translocation
  • Alteration of the gene product to produce transforming proteins
  • Accumulation of molecular alterations in the genome of somatic cells is the basis of cancer progression.

    Figure 1

    Representation of the genomic and histopathological steps associated to tumor progression: from the occurrence of the initiating mutation in the founder cell to metastasis formation. It has been convincingly shown that the genomic landscape of solid tumors such as that of pancreatic and colorectal requires the accumulation of many genetic events, a process which requires decades to complete This timeline offers an incredible window of opportunity for the early detection (often associated to excellent prognosis) of this disease.

    The availability of the human genome sequence and progress in DNA sequencing technologies has dramatically improved knowledge of this disease.

    • The genomic maps are redesigning the tumor taxonomy by moving it from a histologic- to a genetic-based level.
    • The success of cancer drugs designed to target the molecular alterations underlying tumorigenesis has proven that somatic genetic alterations are legitimate targets for therapy.
    • Tumor genotyping is helping clinicians to individualize treatments by matching patients with the best treatment for their tumors.
    • Tumor-specific DNA alterations represent highly sensitive biomarkers for disease detection and monitoring.
    • Finally, the ongoing analyses of multiple cancer genomes will identify additional targets, whose pharmacological exploitation will undoubtedly result in new therapeutic approaches.

 

All dividing cells follow the same basic sequence for replication. The cell generation time is the time required to complete the five phases of the cell cycle.

The G1 phase (G = gap) involves various cellular activities, such as protein synthesis, RNA synthesis, and DNA repair. When prolonged, the cell is considered to be in the G0 phase, that is, the resting phase. G1 cells may either terminally differentiate into the G0 phase or reenter the cell cycle after a period of quiescence.

During the S phase, new DNA is synthesized.

The G2 (premitotic) phase is characterized by cells having twice the DNA content as they prepare for division.

Finally, actual mitosis and chromosomal division takes place during the M phase.

Tumors do not typically have faster generation times, but instead have many more cells in the active phases of replication and have dysfunctional apoptosis, hence proliferation. In contrast, normal tissues have a much larger number of cells in the G0 phase. As a result, cancer cells proceeding through the cell cycle may be more sensitive to chemotherapeutic agents, whereas normal cells in G0 are protected. This growth pattern disparity underlies the effectiveness of chemotherapeutic agents.

In principle, chemotherapeutic drugs are able to treat cancer and spare normal cells by exploiting inherent differences in their individual growth patterns. Each tumor type has its own characteristics, which explain why the same chemotherapy regimen is not equally effective for the whole spectrum of cancers. Selecting appropriate drugs and limiting toxicity demands an understanding of cellular kinetics and biochemistry.

Agents are organized according to the cell cycle stage in which they are most effective for tumor control.

 

 

 

 

 

 

 

 

 

Doubling Time

The time needed for a tumor to double in size is commonly referred to as its doubling time. Whereas the cell cycle generally refers to the activity of individual tumor cells, doubling time refers to the growth of an entire heterogenous tumor mass. In humans, the doubling times of specific tumors vary greatly.

The speed with which tumors grow and double in size is largely regulated by the number of cells that are actively dividing—known as the growth fraction. Typically, only a small percentage of the tumor will have cells that are rapidly proliferating. The remaining cells are in the G0 resting phase. In general, tumors that are cured by chemotherapy are those with a high growth fraction, such as gestational trophoblastic neoplasia. When tumor volume is reduced by surgery or chemotherapy, the remaining tumor cells are theoretically propelled from the G0 phase into the more vulnerable phases of the cell cycle, rendering them susceptible to chemotherapy.

Cell Kinetics

Chemotherapeutic agents typically work by first-order kinetics to kill a constant fraction of cells rather than a constant number. For example, one dose of a cytotoxic drug may result in a few logs (102 to 104) of cell kill. This, however, is not curative since tumor burden may be 1012 cells or more. Thus, the magnitude of cell kill necessary to eradicate a tumor typically requires intermittent courses of chemotherapy. In general, a cancer's curability is inversely proportional to the number of viable tumor cells at the beginning of chemotherapy.

Some drugs achieve cell kill at several phases of the cell cycle. These cell cycle-nonspecific agents act in all phases of replication from G0 to the M phase. Cell cycle-specific agents act only on cells that are in a specific phase. By combining drugs that act in different phases of the cell cycle, the overall cell kill should be enhanced.




 

 

 



Despite epidemiologic evidence implicating diet in digestive tract cancer, the roles of specific dietary elements remain uncertain. Suspected dietary factors in large bowel cancer include excess consumption of animal fat and red meat and excess total caloric intake.52-54 Although low fiber consumption has been suggested to increase colorectal cancer risk, a randomized trial and other recent studies did not show any benefit of a high-fiber, low-fat diet in reducing the recurrence of colorectal adenomas.55-57

Recommended dietary changes to reduce cancer risk include controlling caloric intake, decreasing fat consumption to less than 30% of total daily caloric intake, and increasing consumption of fresh fruits and vegetables.53 These dietary modifications may also reduce the risk of cardiovascular disease.

In general, despite widespread public interest and concern regarding the role of diet in cancer etiology, specific recommendations regarding appropriate dietary recommendations remain elusive. A major contribution in this regard is a monograph published by the World Health Organization that summarizes much of the current knowledge regarding the association between diet and cancer prevention.58 Generally, a diet high in fresh fruits and vegetables and low in red meat seems most prudent. Much interest currently focuses on caloric consumption, physical activity, and energy use and how these factors contribute to obesity and cancer risk [see Obesity and Physical Inactivity, below]. Very recently, attention has focused on early-life patterns of food and caloric consumption and their role in establishing future cancer risk.

Numerous randomized trials have been undertaken to clarify the relationship between diet and cancer. Antioxidant vitamins, minerals such as selenium and calcium, low-fat diets, and other dietary patterns have been assessed as possible risk-lowering factors. Results have differed across these studies, and many studies are ongoing. The Alpha-Tocopherol, Beta-Carotene Cancer Prevention Trial randomized 29,133 Finnish male smokers to receive vitamin E, b-carotene, both, or a placebo to determine whether these supplements lowered the risk of lung cancer. Ironically, the study showed an 18% increased risk of lung cancer in patients who received b-carotene.59 A postintervention follow-up study concluded that smokers should avoid b-carotene supplements.60 These results suggest that dietary interventions may be harmful, and careful investigation is needed to determine their role in cancer prevention.61 In the SELECT trial, a multinational, randomized, and placebo-controlled trial, selenium and vitamin E did not prevent prostate cancer in relatively healthy men.62

Obesity and Physical Inactivity

Two risk factors for cancer that have come under increased study in recent years have been obesity and physical inactivity, which are related to energy intake and consumption. A study by Calle and colleagues demonstrated that obesity, which has been associated most notably with breast cancer and endometrial cancer, may be linked to a larger number of cancers than had previously been suspected, including colon, rectum, prostate, pancreas, kidney, and others.63 An association with adenocarcinoma of the esophagus by increasing GERD has also been fairly consistent. Although the physiologic mechanisms linking obesity to hormonally related cancers (e.g., breast cancer and prostate cancer) are well elucidated, the biologic underpinnings between obesity and other cancers are not obvious.

Physical inactivity, which frequently accompanies obesity, has been implicated as a risk factor in many chronic diseases and is currently being assessed for its contribution to cancer risk. A Canadian study showed that physical inactivity, high energy intake, and obesity are associated with an increased risk of rectal cancer; the study further suggested that these three factors may have a synergistic effect that further increases cancer risk if they are all present.64

Increased physical activity, either on an occupational or a recreational basis, has been shown to be a protective factor for a number of malignancies, including colorectal and breast cancer.65 Whether this represents a surrogate for a healthy lifestyle, whether it serves to counteract some of the deleterious effects of obesity, or whether some other physiologic mechanisms are at work remains unclear. Nonetheless, randomized trials are under way to assess its efficacy as a modifiable intervention for cancer prevention in persons at high risk for developing certain cancers.

Infectious Agents

Infectious agents have been known to cause cancer since 1911, when Peyton Rous discovered the Rous sarcoma virus. However, recent findings have made it clear that infectious agents are responsible for a larger portion of the cancer burden in the United States and around the world than was previously suspected. This is a cause for optimism because it suggests that the preventive and management strategies developed for controlling infectious diseases, including sanitation, vaccination, antibiotics, and antivirals, may be applicable to cancer prevention.

Hepatitis B virus (HBV) infection is an established risk factor for the development of liver cancer in certain patient populations. Cancers of the liver, primarily hepatocellular carcinoma (HCC), are relatively rare in the United States, but the rates are rising.66 HCC occurs largely in patients with cirrhosis caused by chronic hepatitis, hemochromatosis, and the use of exogenous androgenic steroids. It also may occur as a result of occupational exposure to vinyl chloride and the subsequent development of angiosarcoma of the liver. Liver cancer is a leading cause of cancer death in certain developing nations, particularly in Africa and Asia. The major risk factor for liver cancer in these patients is chronic HBV infection, which is often acquired during infancy or early childhood. The risk of liver cancer is increased as much as 200-fold in HBV carriers. Hepatitis B vaccination decreases the risk of infection and subsequent occurrence of HCC. Since the initiation of wide-scale hepatitis B vaccination in Taiwan, HCC rates have fallen by 25% or more.67,68 In the United States, however, the rising incidence of hepatitis C virus (HCV), which causes hepatoma and for which a vaccine does not yet exist, poses an increasing problem.69

Squamous cell carcinoma of the uterine cervix has been linked to human papillomavirus (HPV)70,71 and has been studied in relation to sexual behavior. The incidence of the disease is increased in prostitutes and in other women who have multiple sexual partners, in women who have first coitus at an early age, and in women with a history of venereal disease. Cohort and case-control studies have demonstrated that HPV is associated with both in situ and invasive cervical carcinoma. HPV DNA has been found in up to 95% of cervical cancers. Furthermore, progression of cervical intraepithelial neoplasia is associated with high levels of HPV DNA within the cervical tissues.

This finding has prompted two advances in the prevention and early detection of cervical cancer. First, the presence of HPV in the genital mucosa of women is now being used as a means for cervical cancer screening.72 This technique has proved to be inexpensive and effective and has a high enough sensitivity and specificity for use in poor countries, where the more commonly used Papanicolaou smear is not routinely available. Women are able to swab themselves; the swab is tested for the presence of HPV DNA, and its presence is then used to select those women for further testing where routine gynecologic care is not feasible.73

A second major advance linked to the viral etiology of cervical cancer is the development of a vaccine for HPV. Approximately 70% of cervical cancer is caused by sexual transmission of HPV types 16 and 18. Efforts have been made to develop vaccines that could confer protection against infection by these HPV types, thereby reducing the incidence of preinvasive lesions that eventually lead to invasive cervical cancer. These vaccines are made with viruslike particles that express only the HPV L1 protein but are noninfectious and highly immunogenic. Two products have been studied thus far: a quadrivalent vaccine against HPV types 6, 11, 16, and 18 (types 6 and 11 being responsible for the majority of genital warts) and a bivalent vaccine against HPV types 16 and 18, which may also confer cross-protection against HPV types 31 and 45.74,75 In two randomized trials, both vaccines appeared to be highly effective in preventing HPV infection, conferring protection up to 4.5 years after the administration of three doses over a 6-month period.74,75 The quadrivalent vaccine (Gardasil) was approved by the Food and Drug Administration as a cancer prevention agent for patients 9 to 26 years of age; studies of the bivalent vaccine are ongoing.70

Two phase III studies have confirmed Gardasil to be highly effective in providing protection from cervical cancer. In the first study, the vaccine provided 100% protection from cervical, vaginal, and vulvar diseases caused by HPV types 6, 11, 16, and 18.76 In the second study, Gardasil provided 98% protection against advanced cervical precancerous lesions caused by the two primary cancer-causing HPV types, 16 and 18.77

In addition to cervical cancer, HPV is also implicated in a subgroup of head and neck squamous cell carcinoma (HNSCC), particularly in the oropharynx region, and tends to affect young patients without excessive exposure to tobacco and alcohol.78 Two viral oncogenes, E6 and E7, from HPV type 16 infection, are associated with the malignant transformation.79 HPV-associated HNSCC has a better prognosis and response to treatment compared with HPV-negative HNSCC.80,81

Another virus of interest as a carcinogen is EBV. This DNA virus has been associated with a number of different malignancies, including nasopharyngeal carcinoma, Burkitt lymphoma, posttransplant lymphoproliferative disease, and possibly Hodgkin disease.

Particularly since the early 1980s, with the onset of AIDS and the HIV era, interest has focused on finding retroviruses or RNA viruses associated with cancer. This has been of particular interest because many of the oncogenic viruses found in rodent and animal studies (e.g., SV40 or Rous sarcoma virus) are RNA viruses. A major breakthrough in this regard was the discovery that HHV-8, a retrovirus, was the etiologic agent for Kaposi sarcoma.82 Kaposi sarcoma, which classically occurs in elderly men of Mediterranean descent, became more widespread as a cancer commonly found in AIDS patients. Moore and Chang found HHV-8 to be the virus responsible for Kaposi sarcoma in both the AIDS-associated and the classic forms of the disease.82 HHV-8 has been found to be responsible for Castleman disease and other lymphomas as well.

Human T cell lymphotropic virus type I (HTLV-I) infection has been linked to the development of an aggressive form of leukemia, adult T cell leukemia/lymphoma.83 This disease has an unusual geographic distribution, with clusters in Japan and the Caribbean, and a prolonged latency period. Spatial and temporal clusters of leukemia and Hodgkin disease have also been reported, but no etiologic agent has been found, and these clusters may be the result of chance. HIV type 1 (HIV-1) has been identified as the cause of AIDS. Patients with AIDS are at high risk for Kaposi sarcoma, Hodgkin disease, non-Hodgkin lymphoma (NHL), and other cancers.

In recent years, the incidence of NHL has increased in adolescents and adults. Both inherited and acquired immunodeficiency appear to be associated with an elevated risk of NHL. Organ transplant recipients are at high risk for NHL and carcinomas of the skin.84 These neoplasms may appear within several weeks after renal or cardiac transplantation and therefore differ from most environmentally induced cancers that arise many years after exposure to carcinogens. The transplant-associated lymphomas have a predilection for the central nervous system. Immunosuppressive therapy with azathioprine and cyclosporine has been implicated as a risk factor in transplant recipients, although the transplanted cells per se may also have a carcinogenic influence. EBV has been associated with nasopharyngeal cancer, Burkitt lymphoma, NHL, and Hodgkin disease.12

Evidence accrued over the past 20 years has established H. pylori as a causal agent in gastric cancer. This discovery is significant because it represents the first time a bacterium has been found to be associated with cancer etiology. H. pylori infection is a widely prevalent infection that increases the risk of gastric adenocarcinoma and gastric lymphoma of mucosa-associated lymphoid tissue (MALT) lymphomas.14,15 The treatment of H. pylori infection with antibiotics has resulted in cures of MALT tumors. Furthermore, a recent randomized trial from China demonstrated that early treatment of H. pylori with antibiotics can decrease the subsequent incidence of gastric cancer in persons at high risk for that disease.85

In some parts of the world, such as the Middle East, a large number of cases of bladder cancer are attributable to chronic schistosomiasis, a parasite.

Occupational Carcinogens

Asbestos induces lung cancer and mesothelioma and is a major cause of cancers of the respiratory tract. Each year in the United States, asbestos is estimated to cause several thousand cancer cases, primarily lung cancer.8 Asbestos is virtually indestructible and is widely used throughout industrialized environments. Mesothelioma has been reported to develop after a single identifiable exposure to asbestos. In contrast, lung cancer develops chiefly in workers who have been heavily exposed to asbestos, such as asbestos manufacturers, pipe fitters, and shipyard workers. Concerns have also been raised regarding low-level asbestos exposure associated with decaying insulation in older homes and buildings.86 The carcinogenic effect of asbestos appears to be related to the long, needlelike physical configuration of the fiber. There are concerns that other fibers, such as certain fiberglass insulation materials, might pose a cancer risk.87 Smoking and exposure to asbestos appear to act synergistically in producing cancer. The combination of smoking and occupational exposure to asbestos increases lung cancer risk about 60-fold.

Bladder cancer has been reported in workers exposed to certain aromatic amines in the dye, rubber, leather, tanning, and organic chemical industries. This has been found to be true for kidney cancers as well.

Other occupational carcinogens include polycyclic aromatic hydrocarbons, such as those found in cigarette smoke. These are also derived from combustion of petroleum and its by-products in diesel engines or in other similar settings.

Certain miners, such as uranium miners, can be exposed to unusual carcinogens such as radon. Radon is inhaled and increases the risk of lung cancer. Studies have shown that there is a dose-response relationship associated with mining and that an interactive effect with cigarette smoking increases the risk of lung cancer in these miners.88 Other types of miners, such as coal miners, may also have an increased risk of lung cancer.

Carcinogens Affecting the Reproductive System

Breast cancer is the most common neoplasm in women in the United States and accounts for 28% of all new cancers diagnosed in women in 2010.1 The cumulative incidence of breast cancer in American women is 9% by 74 years of age. The highest rates worldwide are observed in the industrialized countries of North America and Europe. Rising breast cancer rates in Japan and other newly industrialized nations, as well as in Asian immigrants to the United States, suggest environmental influences.7,89

Studies have focused on hormonal factors and the influence of obesity, exercise, diet, and parity in promoting carcinogenesis in the breast and female reproductive organs.90-95 The risk of breast cancer increases with early menarche, nulliparity, older age at first birth, and late menopause. Available data suggest a slight increase in breast cancer risk with prolonged use of oral contraceptives in women younger than 35 years but not in older women.90 Prolonged use of postmenopausal hormone replacement therapy has been shown to increase the risk of breast cancer, and the addition of progesterone appears to exacerbate this effect.91,96 The Women's Health Initiative (WHI) demonstrated that postmenopausal estrogen use increases breast cancer incidence and mortality.97 Following reports of increased breast cancer of postmenopausal hormonal use, decreased breast cancer incidence in the United States is attributed to decreased use of postmenopausal hormonal therapy as a result of the WHI results.98

In addition, oophorectomy reduces the incidence of breast cancer by about 50%. Taken together, these observations support a unified hypothesis that breast cancer development is promoted by prolonged exposure to cyclic ovarian secretion of estrogens and progesterone. Furthermore, low to moderate alcohol use increases the risk of breast cancer, with a strong association with hormone-sensitive breast cancer.99,100

A higher risk of breast cancer has been found in women with benign lesions with histologic evidence of proliferation and atypia. Because most of the known risk factors for breast cancer cannot be modified easily, chemopreventive strategies are being explored for women at high risk for cancer. A large randomized trial has determined that tamoxifen substantially reduces the incidence of breast cancer in women at high risk and reduces the rate of contralateral breast cancer in breast cancer patients.4 A second trial found that raloxifene can also serve as chemoprevention for breast cancer.101 Use of conjugated estrogens during menopause appears to retard the progression of osteoporosis and reduce menopausal symptoms.96 Little is known about environmental causes of carcinoma of the ovary and endometrium. These malignant tumors have some risk factors in common with breast carcinoma, including increased risk with nulliparity and perhaps with obesity and high socioeconomic class.

Multiple primary adenocarcinomas of the breast, ovary, and endometrium have been reported in individuals and in families.8 The risk of both ovarian and endometrial cancer may be lower in women who have used oral contraceptives. For women who were exposed in utero to diethylstilbestrol, the risk that adenocarcinoma of the vagina will develop in their early adult years is 1 in 1,000. Otherwise, this cancer is extraordinarily rare.8

In the past 25 years, age-adjusted incidence rates for prostate cancer in the United States have more than doubled.1 Prostate cancer is estimated to account for 28% of all incident cancers in males in the United States. Much of the increase in incidence can be explained by the widespread use of prostate-specific antigen (PSA) for early cancer detection.102 Clinical prostate cancer is much more common in the United States than in Japan. In both countries, however, a large proportion of elderly men have in situ malignant changes in the prostate. Prostate cancer incidence and mortality have risen more rapidly in African Americans, whose age-adjusted mortality for prostate cancer is more than double that of whites (56.3 versus 23.6 per 100,000 from 1990 to 1996).1 Benign prostatic hyperplasia, a common condition in men older than 60 years, does not appear to be strongly associated with prostate cancer risk.

Environmental influences have been examined as an explanation for the rising incidence of prostate cancer and the higher frequency of advanced prostate cancer in the United States, even in the Asian-American population. Dietary and hormonal influences have been examined in relation to prostate cancer, but their role is still unclear. Evidence suggests that a slightly increased risk of prostate cancer is associated with increased consumption of saturated fat and red meat; vitamins A and D and b-carotene may not decrease the risk.102

Iatrogenic Causes

In more than 50 patients treated with cyclophosphamide for cancer and other serious diseases, bladder cancer developed after cyclophosphamide-induced cystitis.103 A second drug, phenacetin, is associated with cancer of the renal pelvis and bladder.104

Ionizing radiation can induce all major forms of leukemia except chronic lymphocytic leukemia.8 Studies have provided evidence for and against the association between electromagnetic radiation and risk of leukemia; current evidence tends not to support this association.105 A high incidence of acute myeloid leukemia has been reported in cancer patients treated with alkylating agents (i.e., melphalan, cyclophosphamide, chlorambucil, and the nitrosoureas), topoisomerase inhibitors, or epipodophyllotoxins.106 In several large studies, the cumulative leukemia risk at 10 years of follow-up was approximately 5%; the leukemogenic effect of these drugs diminishes with longer follow-up. Clinically, the risk of secondary leukemia needs to be weighed against the benefit of treatment. Alternative nonleukemogenic therapies should be sought; antimetabolites, such as cytarabine, fluorouracil, and methotrexate, do not appear to be carcinogens. Patients who undergo bone marrow transplantation are at higher risk for new solid cancers for at least 10 years after transplantation.107 The use of granulocyte colony-stimulating factor (G-CSF) is associated with increased risk of acute myeloid leukemia or myelodysplastic syndrome.108

Miscellaneous Environmental Causes

Ionizing radiation has been recognized as a human carcinogen for a century. This has been a major etiologic factor for cancer following the atomic bomb in Japan, as well as in various occupational settings. Approximately 3 to 5% of all cancers can be attributed to ionizing radiation, including high-dose therapy for cancer and other diseases.8 Radiation can induce brain tumors, cancers of the skin and thyroid, sarcomas of bone and soft tissue, and cancers of other sites.11 Cancer of the skin of the scrotum occasionally develops in workers topically exposed to soot and mineral oils. Chemical exposures and viruses have been studied as etiologic agents in brain tumors, Hodgkin disease, and multiple myeloma, but definitive results are scanty. Although several reports have suggested an association between electromagnetic radiation and leukemia and brain cancer, more definitive studies have demonstrated no evidence of an association.105 Studies have examined the role of herbicides and defoliants, including Agent Orange, which was heavily used during the war in Vietnam. Agent Orange contains dioxins, which are potent animal carcinogens, although no definitive evidence has been found for increased cancer risk in Vietnam War veterans.109 Excesses of soft tissue sarcoma and NHL have been reported in workers exposed to dioxins through their manufacture or use in farming for long periods.8

Inherited Factors that Predispose to Cancer

Introduction

Interest in hereditary influences emerged with the understanding that genetic alterations underlie the process of transformation of a normal cell into a cancer cell. The basis of cancer is the loss of normal genetic control of cellular processes. At the molecular level, cancer is a disorder of genes, particularly oncogenes and tumor suppressor genes. Furthermore, epigenetic control of gene expression is shown to inactivate tumor suppressor genes in various malignancies. Among the estimated 50,000 genes in the human genome, only a small fraction seems to be essential for cancer development. Genetic alterations can alter regulation of cell replication, DNA repair, apoptosis, and immune surveillance against tumor cells. Inherited traits can interact with environmental carcinogens. For example, sunlight increases skin cancers in genetically susceptible Celts and other light-skinned populations. Patients with albinism or xeroderma pigmentosum have a defect in excision repair of ultraviolet light-damaged DNA and develop multiple skin cancers in exposed areas. Epidemiologic studies can help define the influences of both environmental carcinogens and host factors in cancer development.110

Hereditary factors in cancer development have been identified in studies of families with a history of cancer. Virtually every form of cancer manifests a tendency to aggregate in families. Close relatives of a cancer patient are at increased risk for the same form of cancer and perhaps other cancers. The excess site-specific cancer risk is usually two to three times above the age- and sex-specific population rate. However, germline (inherited) mutations in some cancer genes can increase the likelihood of cancer development to nearly 100%. These potent cancer genes are rare in the population but serve as important models for studies of carcinogenesis.111

High-Penetrance Genetic Factors

Risk Factors for Retinoblastoma

A paradigm of hereditary cancers in humans is retinoblastoma. Approximately one third of this childhood cancer occurs in an autosomal dominant pattern with high penetrance. Carriers of a mutated retinoblastoma gene (Rb-1) have more than a 10,000-fold excess risk of this eye tumor, as well as a marked increase in the risk of second cancers (sarcomas, melanomas, and brain tumors).112 The Rb-1 gene is a tumor suppressor. Loss or inactivation of both alleles of the gene abolishes its tumor suppressor function and leads to tumor development. In hereditary retinoblastoma, one abnormal allele has been inherited from a parent. However, this eye tumor develops only after the second normal allele has been inactivated through a somatic (acquired) mutation or other mechanism. Although inherited mutations in the Rb-1 gene are rare, somatic mutations in the gene are active in the genesis of many forms of cancer, including carcinomas of the breast, the lung, and other sites. The Rb-1 protein interacts with the transcription factors and cyclins that regulate progression through the cell cycle. The Rb-1 gene can also be inactivated through binding by the transforming proteins of several oncogenic viruses; this protein-protein interaction is one molecular mechanism of viral oncogenesis.

Risk Factors for Familial Neoplasms

An increasing number of genetic diseases are associated with a high risk of cancer [see
Table 2].111 Neoplasia is the primary manifestation of some cancer genes, such as the Rb-1 gene. Carriers of these genes can often be identified by a family history of the same cancer in multiple relatives affected at unusually early ages. In other genetic disorders, such as neurofibromatosis, cancer is a less common manifestation of the underlying genetic disease. The finding of a predisposing genetic disorder should alert the physician to the possibility of early diagnosis of associated cancers through periodic surveillance.

Risk Factors for Colorectal Cancer

Colorectal cancer tends to develop within certain families. In the dominantly inherited disorder adenomatous polyposis coli (APC), colonic polyps arise in adolescence, and the lifetime risk of colon cancer is nearly 100%. The APC tumor suppressor gene was isolated in 1991 and shown to be the inherited mutation in all polyposis families. In contrast, familial colon cancer without multiple polyposis may be caused by germline mutations in one of the DNA repair genes: MSH2, MLH1, MSH6, PMS1, or PMS2.113,114 Their phenotypes include familial colorectal cancer and hereditary nonpolyposis colorectal cancer, which accounts for 5 to 10% of these cancers in the United States.115

Risk Factors for Reproductive Cancers

A family history of breast cancer is one of the most consistent risk factors of the neoplasm, particularly in women who have multiple relatives with bilateral premenopausal disease. Hereditary breast cancer accounts for approximately 5% of all breast cancers in the United States. Studies have identified at least five genes predisposing to inherited breast cancer. Clinically, the most important is the BRCA1 gene, located on chromosome 17q.116 A second breast cancer gene, BRCA2, has been found on chromosome 13q.117 BRCA1 and BRCA2 account for most of the hereditary breast cancers in young women; carriers of BRCA1 are also predisposed to ovarian cancer of early onset. Women who are carriers of the BRCA1 mutation have a 50 to 85% probability of developing breast cancer by 70 years of age, and their risk for ovarian cancer is 20 to 50%.118 Corresponding figures for breast and ovarian cancers associated with inherited BRCA2 mutations may be lower. The frequency of inherited BRCA1 and BRCA2 mutations varies widely among populations; it appears to be higher among Ashkenazi Jews, approximately 2% of whom carry mutations of BRCA1 or BRCA2.118

Another susceptibility gene for breast cancer is the p53 tumor suppressor gene, the inherited defect in most families with dominantly inherited Li-Fraumeni syndrome. The PTEN gene, which is associated with Cowden disease, also confers an increased risk of benign and malignant breast tumors, as well as brain, prostate, and thyroid neoplasms.119 Breast cancer genes might include the ataxia-telangiectasia gene, on chromosome 11q. Ataxia-telangiectasia is a rare

[Table 2. Common Hereditary Cancers and Syndromes Attributable to Germline Mutations in Predisposing Genes]

autosomal recessive disease in which homozygotes develop neurologic, neoplastic, and other disorders in childhood.

Prostate cancer has a hereditary component, with male relatives of prostate cancer patients exhibiting a twofold increased risk. Hereditary prostate cancer, which accounts for 5 to 10% of all cases, is primarily associated with early-onset disease. Major susceptibility loci for hereditary prostate cancer were recently mapped on chromosome 1 and the X chromosome.120,121

Other High-Penetrance Genetic Factors

Inherited susceptibility plays a role in other common forms of human cancers, including endocrine and brain tumors, skin cancer, kidney cancer, melanoma, and the hematologic neoplasms. Familial forms of these cancers account for a small fraction of incident cases. Within affected families, however, the inherited cancer gene is an exceptionally potent oncogenic influence that can lead to cancer among multiple relatives.

Low-Penetrance Genetic Factors

In addition to highly penetrant cancer susceptibility genes, many low-penetrance genetic variants (polymorphisms) may interact with environmental agents and other genes to modify cancer risk.19,122 These polymorphisms are associated with only moderate increases in risk, but they can occur at high frequency in the population and contribute to the development of substantial numbers of cancers.17,18,20 Unfortunately, the effects of these variants are often small and difficult to measure in heterogeneous populations. For example, rare HRAS1 alleles in the H-ras-1 oncogene are reportedly associated with increased breast cancer risk, but new results based on improved analytical methods have failed to support the finding.122,123 Inconsistent results in studies of low-penetrance cancer genes might also be the result of small sample sizes and misclassification of carcinogenic exposures.124 Characterization of the role of genetic variants in cancer development can help identify susceptible populations to individualize cancer prevention efforts.

Polymorphisms in genes that encode for proteins involved in metabolism of steroid hormones or carcinogens might alter cancer risks. Variants in the cytochrome P-450 genes are associated with increased risk of lung, esophageal, and head and neck cancers. The genes GSTM1 and GSTT1 produce glutathione transferases that are involved in the deactivation of tobacco carcinogens.17 Certain variants in these genes are associated with an increased risk of bladder cancer and a lower survival rate for patients with lung cancer.17 A combination of genetic variants in both cytochrome P-450 genes and GSTM1 may produce gene-gene interactions that further increase cancer risks.18 Polymorphisms of the NAT1 and NAT2 genes may affect susceptibility to cancers of the urinary bladder, colon and rectum, breast, head and neck, and lung.19,20 Individuals with the slow NAT1 and NAT2 acetylator phenotypes may be prone to bladder cancer, whereas those with fast acetylator phenotypes may be predisposed to colorectal cancer.20

Before familial cancers are attributed to genetic susceptibility, chance association and shared exposures to environmental carcinogens should be excluded. Inherited predisposition can be identified through detection of laboratory markers of host susceptibility and by segregation and linkage analysis of the pedigree. With the increasing identification of cancer susceptibility genes, genetic testing of individuals is becoming more widespread. Analysis of the Rb-1 gene has been used to detect carriers in families with retinoblastoma. In affected families, surveillance of newborns for early cancer can reduce loss of vision. As genes for more common cancers have been found, ethical and social issues have become more complex. Careful consideration must be given to the costs and benefits of genetic testing.125

Epigenetics

The control of gene expression is not simply by alterations in the DNA sequence; it is also controlled through modifications of the DNA sequence and the regulatory proteins. The study of epigenetics initially found that global hypomethylation of DNA was increased in human cancers. More hypermethylated tumor suppressor genes were discovered, as well as the inactivation of microRNA (miRNA) genes by DNA methylation. Both histone deacetylation and DNA methylation have been shown to repress gene expression.126 Methylation is a marker of an inactive gene, and the methylation of the promoter region of the MLH1 gene results in colon cancer. VHL gene hypermethylation and the development of renal cell carcinoma have also been shown. However, hypomethylating agents, such as azacytidine, do not have significant activity in solid malignancies; they are shown to increase overall survival and progression-free survival in myelodysplastic syndrome.
Screening and Early Detection

The recommendations of the American Cancer Society, the National Cancer Institute, and the U.S. Preventive Services Task Force for the prevention and early detection of cancer have received wide attention. These guidelines are intended primarily for asymptomatic patients at average risk for cancer. They do not apply to symptomatic patients, who should be managed by the usual standards of medical practice. Patients considered to be at increased risk because of family history or environmental exposures should seek a physician's recommendations for establishing an appropriate early detection program.

Data are incomplete regarding the costs, risks, and benefits of cancer screening in the general population. A few large randomized studies have been completed for specific cancers, such as breast cancer. These studies have found that periodic mammography reduces breast cancer mortality by 20 to 25% in women 50 to 70 years of age. Data are scanty and uncertain for younger and older women.128 To be useful, a screening test must detect preclinical cancers that are less likely to be lethal after treatment than if they are allowed to progress to clinical detection. False positive results of screening tests lead to unnecessary workups and treatment for indolent tumors and increase medical, psychological, and financial costs.129 The identification of high-risk populations through risk evaluation, including genetic testing, can help channel scarce resources to susceptible persons.

The American Cancer Society has made specific recommendations for the early detection of asymptomatic carcinoma of the colon and rectum, cervix and other pelvic organs, breast, and prostate [see
Table 3].130 However, because of the lack of convincing evidence of benefit from screening for lung cancer, periodic chest x-rays and sputum cytology are not recommended, nor are computed tomographic (CT) scans. Recent results from the National Lung Screening Trial (NLST) from the National Cancer Institute report a 20% reduction in lung cancer mortality for smokers undergoing low-dose CT chest scans versus chest x-rays.131 This new finding may have an impact on lung cancer screening in coming years. PSA is being increasingly used for prostate cancer detection. Serum PSA levels correlate with the clinical stage of prostate cancer and the volume of the cancer in the gland.132 Evidence regarding prostate cancer screening with PSA and digital rectal examination is conflicting. The US Prostate, Lung, Colorectal and Ovarian (PLCO) screening trial found no difference in the rate of death, whereas a European study concluded that the rate of death from prostate cancer was reduced by 20%.133,134 Hence, the National Cancer Institute currently recommends informed decision making between the physician and the patient.

The current screening guidelines can be expected to evolve with the accrual of knowledge and technological advances. For example, the technical quality of mammograms and their interpretation are critical factors in their usefulness. Mammographic detection of cancer is more problematic in the dense breast tissue of younger women, whose tumor growth rates are generally more rapid. After analyzing the same body of evidence from randomized mammographic studies, expert committees have reached different conclusions regarding routine mammographic screening of asymptomatic women 40 to 49 years of age. The American Cancer Society, the National Cancer Institute, and other groups have recommended that women begin receiving annual mammograms once they reach 40 years of age, but the National Institutes of Health consensus panel and the U.S. Preventive Services Task Force did not make this recommendation.130,135 Of several methods available for colorectal cancer screening, colonoscopy was found to be superior to both double-contrast barium enema and sigmoidoscopy in two studies.136,137 Of note, CT colonography every 5 years has been added to recommendations from the American Cancer Society.130 New screening methods, such as magnetic resonance imaging of the breast and spiral CT of the lungs, may further enhance the sensitivity and specificity of screening tests.138

Clinicians can help prevent and detect cancer at early stages. They can prevent environmental cancers by counseling patients to avoid tobacco use, asbestos exposure, and unnecessary exposure to ionizing radiation. A brief medical and family history can reveal an unusual predisposition to cancer and a need for closer medical surveillance. In the course of the physical examination, attention to signs of early cancer can lead to curative treatment.

[Table 3. American Cancer Society Recommendations for Early Cancer Detection]

Diet

Unusual geographic clustering has been observed for certain cancers of the digestive tract. Exceptionally high mortality rates are found for esophageal cancer in southern Africa and in parts of China and for stomach cancer in eastern Asia and eastern Europe. In the United States, nearly one fifth of all cancers arise in the digestive tract, most often in the colon or rectum. For unknown reasons, cancers of the right colon have become more common, whereas sigmoid colon and rectal cancers have diminished. Death rates from stomach cancer in the United States have declined sharply over the past 70 years, possibly because of improvements in food preservation and decreased consumption of salted, pickled, and smoked foods.50 Incidence rates of certain digestive tract cancers change when people migrate and, as a result, modify their diets. For example, Japanese who migrate to the United States have a decline in stomach cancer rates, but their colon cancer rates rise within one generation to the levels found in the American-born white population.50,51 Within the United States, colon cancer rates differ among subpopulations. Mormons and Seventh-Day Adventists, who tend to consume less meat, have lower rates of large bowel cancer. Mexican Americans have low rates of bowel cancer but high rates of stomach cancer.

 

 

Cancer Cell Growth

Tumors are characterized by a gompertzian growth pattern (Fig. 27-2). Fundamentally, a tumor mass requires progressively longer times to double in size as it enlarges. When a cancer is microscopic and nonpalpable, growth is exponential. However, as a tumor enlarges, the number of its cells undergoing replication decreases due to limitations in blood supply and increasing interstitial pressure.

 
The gompertzian growth curve. During early stages of tumor expansion, growth is exponential, but with enlargement, tumor growth slows. Consequently, most tumors have completed their exponential growth phase at the time of clinical detection.

When tumors are in the exponential phase of gompertzian growth, they should be more sensitive to chemotherapy because a larger percentage of cells are in the active phase of the cell cycle. For this reason, metastases should be more sensitive to chemotherapy than the primary tumor. To capitalize on this potential benefit, advanced ovarian cancer is usually first treated with surgery to remove the primary tumor, debulk large masses, and leave only microscopic residual disease for the adjuvant chemotherapy to act upon. In addition, when a tumor mass shrinks in response to treatment, the presumption is that a greater number of cells will enter the active phase of the cell cycle to accelerate growth. This larger percentage of replicating cells should also increase the sensitivity of a tumor to chemotherapy.

 

Helicobacter pylorus

The bacterial pathogen Helicobacter pylorus, a common cause of gastric ulcer, is considered a major factor in the development of human gastric cancer.

Probing head and neck cancer microbiomes

Microbiome and mycobiome (fungal community) of head and neck squamous cell carcinomas (HNSCC).

Our pilot data reveal clear microbiomic differences between HNSCC and matched normal oral epithelium. Tantalizingly, the HNSCC microbiomic differences were associated with regional pathologic nodal status (pN2) and smoking, but not the presence or absence of human papillomavirus or alcohol consumption.

We are now analyzing a large independent series of HNSCC and their oral washes for the microbiome and mycobiome. Mycobiomic analyses are being performed by Mahmoud Ghannoum, PhD, FIDSA, Director of Medical Mycology at Case Western Reserve University School of Medicine and University Hospitals Case Medical Center. Head and neck oncologic surgeons led by Brian Burkey, MD, Vice Chair of Cleveland Clinic’s Head & Neck Institute, and anatomic pathologists led by Lisa Yerian, MD, Cleveland Clinic’s Medical Director of Continuous Improvement and former Section Head of Surgical Pathology, play vital roles in ensuring rigorous histologic accuracy and sterile sampling.

A microbiome role in breast cancer?

The oropharynx is typically colonized by microbes. But does an “occult” microbiome exist in organs such as the breast that are typically considered sterile? Stephen Grobmyer, MD, Cleveland Clinic’s Director of Breast Surgical Oncology and Co-Director of the Comprehensive Breast Cancer Program, and I co-lead a multidisciplinary team examining the microbiome in breast cancer. Other Cleveland Clinic members of this team include Holly Pederson, MD, Director of Medical Breast Services; Benjamin Calhoun, MD, PhD, Director of Breast Pathology; and Charles D. Sturgis, MD, a cytopathologist and staff member of the Department of Pathology.

The breast cancer microbiome team is interested in examining whether and how the microbiome plays a role in promoting, or preventing, breast cancer. They seek answers to such questions as whether microbiomic differences can help sort out prognosis in Oncotype DX® Breast Cancer Assay mid-scores and/or help the breast surgeon determine which breast lesions classed as BI-RADS 3 or 4 (mammographically indeterminate) should be biopsied.

David Serre, PhD, an associate staff member of Cleveland Clinic’s Genomic Medicine Institute, and collaborators at the Case Comprehensive Cancer Center are also interrogating the gut microbiome to see whether it can predict which colonic polyps progress to colon cancer.

Future directions: Biomarkers and therapy targets

If elements of the microbiome play key roles in carcinogenesis, then they can serve as novel biomarkers for progression and prognosis as well as novel targets for therapy. Probiotics or antibiotics could act as adjunctive treatment strategies or even as prevention.

Dr. Eng is the founding Chair of Cleveland Clinic’s Genomic Medicine Institute, Director of the Center for Personalized Genetic Healthcare, and a staff member of Taussig Cancer Institute’s Department of Hematology and Medical Oncology. She holds the Sondra J. and Stephen R. Hardis Endowed Chair of Cancer Genomic Medicine and is a Professor of Molecular Medicine at Cleveland Clinic Lerner College of Medicine. She can be reached at engc@ccf.org or 216.444.3440.

microbiome-chart_550x400

Figure. Microbial subpopulational content at the phylum level of head and neck squamous cell carcinoma (HNSCC) and matched normal mucosa. Each bar represents a sample, and the bars are split to show the microbial content quantitatively of that sample at the phylum level.

From Bebek G, Bennett KL, Funchain P, Campbell R, Seth R, Scharpf J, Burkey B,
Eng C. Microbiomic subprofiles and MDR1 promoter methylation in head and neck
squamous cell carcinoma. Hum Mol Genet. 2012 Apr 1;21(7):1557-1565. Used with
permission from Oxford University Press.

 


 

 

    human papillomavirus with cervical cancer, and hepatitis B and C viruses with liver cancer.

    Hepatitis B and Hepatitis C are associated with the development of hepatocellular cancer; HIV is associated with Hodgkin's and non-Hodgkin's lymphomas; human papillomavirus is associated with cervical cancer and head and neck cancer; and Ebstein-Barr virus is associated with nasopharyngeal cancer. Expression of virus-induced neoplasia may also depend on additional host and environmental factors that modulate the transformation process. Cellular genes are known that are homologous to the transforming genes of the retroviruses, a family of RNA viruses, and induce oncogenic transformation. These mammalian cellular genes, known as oncogenes, have been shown to code for specific growth factors and their corresponding receptors. These genes may be amplified (increased number of gene copies) or mutated, both of which can lead to constitutive overexpression in malignant cells. The bcl-2 family of genes represents a series of pro-survival genes that promotes survival by directly inhibiting apoptosis, a key pathway of programmed cell death.

    a. DNA Viruses

    The infection of cells with an oncogenic DNA virus may result either in productive lytic infection with cell death and release of newly formed virus or in cell transformation to the neoplastic state with little or no virus production but with integration of viral genetic information into the cell DNA.

    Papovavirus

    The papovavirus family consists of: the papilloma viruses of cattle, cottontail rabbit, and humans; the polyomavirus of the mouse; and the simian virus 40, originally termed simian vacuolating agent. The genomes of polyomavirus and SV40 are double-stranded circular DNA molecules with sizes of about 5 kilobase pairs (kb) and comprise two chief groups of genes which are associated with early and late events in the replication cycle. The "early" genes are transcribed soon after infection of a cell and are involved in producing "functional" proteins which participate in viral DNA synthesis but are not found in the virions themselves. The "late" genes encode structural proteins of the viral coat and capsid. In productive lytic infection by these viruses, in permissive hosts, early proteins are formed but then disappear and the structural proteins are assembled into viral particles. In non-permissive cells, derived from an animal species which is not a natural host of the virus, neoplastic transformation can occur. When stable transformation does take place, viral DNA is inserted or "integrated" into the cellular chromosomal DNA; some of the early proteins are persistently synthesized; and viral particles are not produced.

    The early proteins found in tumors induced by polyomavirus and SV40 are termed "T" (for tumor) antigens. Polyomavirus produces three (large, middle, small) T antigens, of which middle T antigen is necessary for transformation. This early protein is bound to the plasma membrane of transformed cells. SV40 produces only two (large, small) T antigens. SV40 large T antigen maintains the transformed state.

    Adenoviruses are found in many species of animals, including humans. The genomes of adenoviruses are double-stranded linear DNA molecules with sizes of about 35-40 kb. In cells transformed by oncogenic adenoviruses, a region of the genome encoding early proteins is always present, and a variety of early proteins can be detected. Among them, the major E1a proteins function in the initiation and maintenance of transformation.

    The genomes of herpesviruses are double-stranded linear DNA molecules with sizes in the range of 140-170 kb. The initiation of transformation by oncogenic herpesviruses appears to be related to the presence of certain DNA sequences although no single T antigen is found.

    In summary, the mechanisms of DNA virus oncogenesis indicate that oncogenesis is an attribute of the viral DNA, that the viral DNA is integrated into the host cell DNA, and that the protein products of viral genes maintain transformation to the neoplastic state.

    DNA viruses, along with other environmental and hereditary factors, are associated with the etiology of several types of human cancer. The Epstein-Barr virus (EBV), a type of herpesvirus and the cause of infectious mononucleosis, may be involved in the causation of Burkitt's lymphoma in Africa and sporadic cases elsewhere, as well as nasopharyngeal carcinoma, and viral DNA and various EBV-determined antigens are detectable in the tumor cells. Hepatitis B virus (HBV) is considered to have a causal role in primary hepatocellular carcinoma, one of the most common forms of cancer in Asia and worldwide, and viral DNA is integrated into the tumor cells in some cases. Hepatitis C virus (HCV) is similarly involved in hepatic carcinogenesis. A newly recognized human herpes virus, first described in 1994 and now designated HHV type 8 or KSHV (Kaposi sarcoma-associated herpes virus), is implicated as a candidate etiologic agent in AIDS-associated KS, the most common malignant tumor seen in patients with acquired immunodeficiency syndrome, as well as in classic (sporadic) KSV unrelated to AIDS and in AIDS-associated B-cell lymphoma of body cavities (primary effusion lymphoma). Some types of sexually transmitted human papilloma viruses (HPV) are associated with precursor lesions of squamous carcinoma of the uterine cervix. HPV viral DNA is extrachromosomal in the precursor lesions, and infectious virus is produced. HPV types 16 and 18 are associated both with precursor lesions and with invasive cervical carcinoma. Viral DNA is integrated into the cancer cells in many studied cases, but additional agents or factors may be involved at different stages of the progression to invasive carcinoma.

    RNA Viruses

    Of the many families of RNA viruses, only members of the retrovirus family are capable of inducing animal tumors and transforming cultured cells. The oncogenic RNA viruses (RNA tumor viruses) are a subfamily of the retroviruses and are a cause of naturally occurring tumors and leukemias in a wide range of vertebrate animals, including mammalian, avian, and reptilian species. The RNA tumor viruses are classified according to their natural host, such as avian, murine, feline, and primate leukemia/ sarcoma virus species. Two unique types of human retroviruses, human T-cell leukemia viruses (HTLV) types 1 and 2 are etiologically associated with human leukemias.Human immunodeficiency virus (HIV), a member of the lentivirus subfamily of retroviruses and the causative agent of AIDS, predisposes to opportunistic infections, including those caused by tumor-related viruses such as KSHV and HPVs.

    The genomes of retroviruses are diploid and are single-stranded RNA molecules with a size range of 3-9 kb. All retroviruses contain a reverse transcriptase (RNA-directed DNA-polymerase), and their replication requires the synthesis of a double-stranded DNA intermediate of the RNA genome. Some of the virally determined DNA becomes inserted, or integrated, into the host cell DNA as provirus DNA. Typically, there are three retroviral genes which encode proteins necessary for viral replication: gag (group-specific antigen) gene which encodes internal structural proteins of the virus; pol gene which encode reverse transcriptase;and env gene which encodes envelope proteins that enclose the virus particles and largely determine the host range.

    Most oncogenic retroviruses also have another gene known as a transforming gene or oncogene and termed v-onc (and usually identified by a 3-letter code, such as src in the prototype Rous sarcoma virus). Under the influence of the viral promoter sequence, the v-onc gene is transcribed along with other viral genes and is responsible for neoplastic transformation of the cell. All rapidly transforming retroviruses possess one, or rarely two, unique oncogenes of which more than 20 have been isolated and characterized. A short list of retroviral oncogenes is given in the table.

    Table: Retroviral Oncogenes (partial list)

     

    Oncogene (v-onc) Prototype Retrovirus Species of Origin

    src Rous sarcoma virus Chicken
    myc Avian myelocytomatosis virus Chicken
    erb A, erb B Avian erythroblastosis virus Chicken
    myb Avian myeloblastosis virus Chicken
    H-ras Harvey rat sarcoma virus Rat
    K-ras Kirsten murine sarcoma virus Mouse
    abl Abelson murine leukemia virus Mouse
    fes Feline sarcoma virus Cat
    sis Simian sarcoma virus Monkey

 

Malignant Transformation of Hymenolepis nana in a Human Host

Atis Muehlenbachs, M.D., Ph.D., Julu Bhatnagar, Ph.D., Carlos A. Agudelo, M.D., Alicia Hidron, M.D., Mark L. Eberhard, Ph.D., Blaine A. Mathison, B.S.M.(A.S.C.P.), Michael A. Frace, Ph.D., Akira Ito, Ph.D., Maureen G. Metcalfe, M.S., Dominique C. Rollin, M.D., Govinda S. Visvesvara, Ph.D., Cau D. Pham, Ph.D., Tara L. Jones, Ph.D., Patricia W. Greer, M.T., Alejandro Vélez Hoyos, M.D., Peter D. Olson, Ph.D., Lucy R. Diazgranados, M.D., and Sherif R. Zaki, M.D., Ph.D.

N Engl J Med 2015; 373:1845-1852November 5, 2015DOI: 10.1056/NEJMoa1505892

 

 

 

azo dyes, aflatoxins, asbestos, benzene, and radon

Tobacco Smoke

Chemical carcinogenesis by tobacco smoke products is a major cause of common lung cancers.

    The discovery of chemical carcinogenesis was made by Sir Percival Pott (1713-1788), English surgeon, who related the cause of scrotal skin cancer in a number of his patients to a common history of occupational exposure to large amounts of coal soot as chimney sweepers when they were boys.

    The table lists chemical agents that have been established over time as causes of occupational cancers.

    Table:

    Agent Occupation Cancer Site

    Polycyclic hydrocarbons in soot, tar, oil chimney sweepers, manufacturers of coal gas, many other groups of exposed industrial workers scrotum, skin, bronchus
    2-Naphthylamine; 1-naph-thylamine chemical workers, rubber workers, manufacturers of coal gas bladder
    Benzidine; 4-aminobiphenyl chemical workers bladder
    Asbestos asbestos workers, shipyard and insulation workers bronchus, pleura, and peritoneum
    Arsenic sheep dip manufacturers, gold miners, some vineyard workers and ore smelters skin and bronchus
    Bis(chloromethyl) ether makers of ion-exchange resins bronchus
    Benzene workers with glues, varnishes, etc. marrow (leukemia)
    Mustard gas poison gas makers bronchus, larynx, nasal sinuses
    Vinyl chloride PVC manufacturers liver (angio-sarcoma)

    Osteogenic sarcoma of femur

    An occupational cancer in a radium dial painter. Neoplastic osteocytes embedded in malignant osteoid matrix (the homogeneous substance between tumor cells).

     

     

    In addition to occupational exposure to carcinogens, medical treatment with agents such as ionizing radiations and natural exposure to solar ultraviolet radiation were early recognized as causes of human cancers. Occupational cancers comprise a small but preventable part of the worldwide incidence of human cancers. It is paradoxical that, more than two centuries after the discovery of the carcinogenic hazards of coal soot, a smoke product of another source, associated with life-style rather than occupation, is etiologically related to one of the most prevalent human cancers today, namely, bronchogenic carcinoma of tobacco smokers.

     

     

     

    Classification of Chemical Carcinogens

    Chemical carcinogens are of synthetic ("man made") or natural origin, are extremely diverse in structure without any common feature, and are classified into two categories:

    • direct-acting (DNA-reactive, activation independent, genotoxic) carcinogens that bind covalently to cellular genomic DNA and are mutagens;
    • procarcinogens (activation dependent) that require metabolic conversion to metabolites ("ultimate carcinogens") capable of transforming cells and inducing tumors.

    Procarcinogens are among the most potent chemical carcinogens.

    Table: Major Chemical Carcinogens


    Pro-carcinogens (require metabolic activation to "ultimate carcinogens")

      Polycyclic aromatic hydrocarbons
      • benzanthracene (first pure carcinogen)
      • 3,4-benzpyrene (isolated from coal tar)
      • 3-methylcholanthrene (prepared from a steroid, deoxycholic acid)
      • 7,12-dimethylbenzanthracene (most potent carcinogen)

      Aromatic Amines and Azo Dyes

      • 2-naphtylamine (produces bladder carcinoma)
      • benzidine (produces bladder carcinoma)
      • 2-acetylaminofluorene
      • 4-dimethylaminoazobenzene (produces liver tumors)

      Natural Products

      • aflatoxin B1 (potent hepatocarcinogen produced by mold contamination of food)
      • mitomycin C

      Other

      • nitrosamine (can be formed by action of nitrites on foods)
      • some insecticides (chlordane and others)
      • some metals (chromium and nickel)
      • carbon tetrachloride

    Direct-Acting Carcinogens (DNA-reactive)

      Alkylating agents
      • anticancer chemotherapeutic drugs (cyclophosphamide, busulfan, chlorambucil)
      • beta-propiolactone
      • bis(chloromethyl)ether

      Acetylating agents

      • 1-acetylimidazole

    Biological Aspects of Chemical Carcinogenesis

    Carcinogenesis is a multiple step process. One of the characteristics of chemical or physical carcinogenesis is the usually extended period of time (latent period) between contact with the carcinogen and the appearance of a tumor. The latent periods of occupational cancers may extend from one to several years and commonly to several decades, as noted in the accompanying table.

    Table: Latent Periods of Representative Occupational Cancers

    LatentPeriod (years)
    Site of Cancer Type of Cancer Agent Average Range

    Skin Epidermoid and basal cell carcinomas Arsenic
    Coal tar & pitch
    Ionizing radiation
    Solar radiation
    25
    20-24
    7
    20-30
    4-46
    1-50
    1-12
    15-40
    Lung Bronchogenic carcinoma Asbestos
    Ionizing radiation
    18
    25-35
    15-48
    7-50
    Bone marrow Leukemia Benzene
    Ionizing radiation
    3-19
    3-15
    Bladder Squamous cell carcinoma Aromatic amines 11-15 2-40
    Bone Osteogenic sarcoma Ionizing radiation 10-25

    Biochemical Aspects of Chemical Carcinogenesis

    Most chemical carcinogens are , or are metabolically converted into, electrophilic reactants (electron-attracting chemicals) that cause their biological effects by covalent binding with cellular proteins and nucleic acids, particularly chromosomal DNA. The most frequent reaction sites in DNA are with guanine.

    The majority of chemical carcinogens are procarcinogens and require metabolic conversion into chemically reactive forms (ultimate carcinogens). Many chemical pathways (oxidation, reduction, hydroxylation, hydrolysis, conjugation, etc.) lead to metabolic conversion of procarcinogens to intermediate metabolites (proximate carcinogens) and finally to ultimate carcinogens which react with cellular DNA to cause neoplastic transformation.

    Most procarcinogens are activated by microsomal enzymes in the endoplasmic reticulum. The conversion of polycyclic aromatic hydrocarbons to ultimate carcinogens is initiated by aryl hydrocarbon hydroxylase (AHH). Cytochrome P-450, a terminal component of an electron transport system present in liver microsomes, is also involved in the metabolic activation of procarcinogens.

    Tests for mutagenicity indicate that virtually all ultimate carcinogens are mutagenic. Conversely, most mutagens show carcinogenic activity. In the Ames screening test for mutagenicity, a putative carcinogen is incubated with liver microsomes and an indicator microorganism. An increase in the frequency of specific mutants above control levels is scored as a positive result .

    Modifying Factors in Carcinogenesis

    Host factors (genetics, gender, hormones, aging) and environmental factors may have a modifying role in increasing, or decreasing, the susceptibility to carcinogens. With procarcinogens, activating enzyme systems must be present or inducible in target cells. This genetically determined activity explains the organ and species specificity of some procarcinogens.

    Microsomal enzymes in the liver degrade (detoxify) a large part of a procarcinogen to non-carcinogenic products. Enzymes can be induced which accelerate detoxification. A variety of naturally occurring compounds, such as indole, flavones, and related compounds that occur in vegetables (brussels sprouts, cabbage, broccoli, cauliflower) have a protective action in animals exposed to carcinogenic polycyclic hydrocarbons.

    Endogenous (and exogenous) sex hormones are important factors apparently in the promotion stage of human carcinomas of breast, endometrium, and prostate. Additionally, a history of exposure in-utero to the synthetic estrogen diethylstilbestrol (DES) is strongly associated with the development of carcinoma of the vagina and cervix in some young women.

    Other exogenous factors in human carcinogenesis include dietary excesses and deficiencies and, most notably, tobacco smoking which is a major factor associated with lung carcinoma.

    Non-genotoxic Carcinogens

    The actions previously described are those of agents which react with cellular DNA and cause genomic alterations. As more and more chemicals are tested for carcinogenicity, a number are now being recognized as "non-genotoxic". These chemicals do not form stable covalent bonds with cellular DNA or other macromolecules. Solid state materials (asbestos) are an example.

    DNA Damage and Repair

    Ultimate carcinogens are mutagens and cause point mutations (base-pair substitutions) and frame-shift mutations. The activation of ras proto-oncogenes by point mutations is associated with chemical carcinogenesis in experimental animals and with many types of human cancers. Ultraviolet and ionizing radiations are also mutagenic and produce several types of lesions (strand breaks, cross-links, base alterations) in cellular DNA. Ultraviolet radiation also produces dimers between adjacent thymidines.

    Excision repair of damaged DNA occurs to a greater or lesser extent and involves the excision of the damaged strand, synthesis of a patch, and rejoining of the strand by a DNA ligase.

    Several autosomal recessive disorders associated with an increased incidence of cancer exhibit defects in the repair and maintenance of DNA. Notably, the repair of damage caused by ultraviolet radiation is defective in patients with xeroderma pigmentosa (autosomal recessive) who have a high incidence of skin cancer in areas exposed to sunlight. Bloom's syndrome, a condition characterized by multiple chromosomal breaks and a high incidence of leukemia or intestinal cancer, is associated with a defect in a DNA ligase.

 

Agent Occupation Cancer Site

Ionizing radiations
radon certain underground miners (uranium, fluorspar,etc.) bronchus
X-rays, radium radiologists, radiographers skin
radium luminous dial painters bone  to 1491
Ultraviolet radiation farmers, sailors, etc. skin

Exposure to ionizing radiation has been well documented as a significant risk factor for a number of cancers, including acute leukemias, thyroid cancer, breast cancer, lung cancer, soft tissue sarcoma, and basal cell and squamous cell skin cancers.

 

 

Specific causes of common human cancers - breast, colon, rectum, lymph nodes, uterus, bladder, pancreas, bone marrow, and stomach, remain unknown.

USMLE Reviewer (Subscription Required)