Top Banner
CHAPTER ONE Molecular Biology of Cancer JESSE D. MARTINEZ MICHELE TAYLOR PARKER KIMBERLY E. FULTZ NATALIA A. IGNATENKO EUGENE W. GERNER Departments of Radiation Oncology/Cancer Biology Section Molecular and Cellular Biology Biochemistry and Molecular Biophysics Cancer Biology Graduate Program The University of Arizona Tuscon, Arizona Burger’s Medicinal Chemistry and Drug Discovery Sixth Edition, Volume 5: Chemotherapeutic Agents Edited by Donald J. Abraham ISBN 0-471-37031-2 © 2003 John Wiley & Sons, Inc. Contents 1 Introduction, 2 2 Tumorigenesis, 2 2.1 Normal-Precancer-Cancer Sequence, 2 2.2 Carcinogenesis, 3 2.3 Genetic Variability and Other Modifiers of Tumorigenesis, 5 2.3.1 Genetic Variability Affecting Cancer, 5 2.3.2 Genetic Variability in c-myc–Dependent Expression of Ornithine Decarboxylase, 7 2.4 Epigenetic Changes, 7 3 Molecular Basis of Cancer Phenotypes, 10 3.1 Immortality, 10 3.2 Decreased Dependence on Growth Factors to Support Proliferation, 11 3.3 Loss of Anchorage-Dependent Growth and Altered Cell Adhesion, 12 3.4 Cell Cycle and Loss of Cell Cycle Control, 14 3.5 Apoptosis and Reduced Sensitivity to Apoptosis, 16 3.6 Increased Genetic Instability, 19 3.7 Angiogenesis, 20 4 Cancer-Related Genes, 21 4.1 Oncogenes, 21 4.1.1 Growth Factors and Growth Factor Receptors, 21 4.1.2 G Proteins, 23 4.1.3 Serine/Threonine Kinases, 24 4.1.4 Nonreceptor Tyrosine Kinases, 24 4.1.5 Transcription Factors as Oncogenes, 25 4.1.6 Cytoplasmic Proteins, 26 1
50

Molecular Biology of Cancer - UC · PDF fileDepartments of Radiation Oncology/Cancer Biology Section Molecular and Cellular Biology ... 2 Molecular Biology of Cancer. are genetic mutations

Mar 17, 2018

Download

Documents

trinhbao
Welcome message from author
This document is posted to help you gain knowledge. Please leave a comment to let me know what you think about it! Share it to your friends and learn new things together.
Transcript
Page 1: Molecular Biology of Cancer - UC · PDF fileDepartments of Radiation Oncology/Cancer Biology Section Molecular and Cellular Biology ... 2 Molecular Biology of Cancer. are genetic mutations

CHAPTER ONE

Molecular Biology of Cancer

JESSE D. MARTINEZ

MICHELE TAYLOR PARKER

KIMBERLY E. FULTZ

NATALIA A. IGNATENKO

EUGENE W. GERNER

Departments of Radiation Oncology/Cancer Biology SectionMolecular and Cellular BiologyBiochemistry and Molecular BiophysicsCancer Biology Graduate ProgramThe University of ArizonaTuscon, Arizona

Burger’s Medicinal Chemistry and Drug DiscoverySixth Edition, Volume 5: Chemotherapeutic AgentsEdited by Donald J. AbrahamISBN 0-471-37031-2 © 2003 John Wiley & Sons, Inc.

Contents

1 Introduction, 22 Tumorigenesis, 2

2.1 Normal-Precancer-Cancer Sequence, 22.2 Carcinogenesis, 32.3 Genetic Variability and Other Modifiers of

Tumorigenesis, 52.3.1 Genetic Variability Affecting Cancer, 52.3.2 Genetic Variability in

c-myc–Dependent Expression ofOrnithine Decarboxylase, 7

2.4 Epigenetic Changes, 73 Molecular Basis of Cancer Phenotypes, 10

3.1 Immortality, 103.2 Decreased Dependence on Growth Factors to

Support Proliferation, 113.3 Loss of Anchorage-Dependent Growth and

Altered Cell Adhesion, 123.4 Cell Cycle and Loss of Cell Cycle Control, 143.5 Apoptosis and Reduced Sensitivity to

Apoptosis, 163.6 Increased Genetic Instability, 193.7 Angiogenesis, 20

4 Cancer-Related Genes, 214.1 Oncogenes, 21

4.1.1 Growth Factors and Growth FactorReceptors, 21

4.1.2 G Proteins, 234.1.3 Serine/Threonine Kinases, 244.1.4 Nonreceptor Tyrosine Kinases, 244.1.5 Transcription Factors as Oncogenes,

254.1.6 Cytoplasmic Proteins, 26

1

Page 2: Molecular Biology of Cancer - UC · PDF fileDepartments of Radiation Oncology/Cancer Biology Section Molecular and Cellular Biology ... 2 Molecular Biology of Cancer. are genetic mutations

4.2 Tumor Suppressor Genes, 264.2.1 Retinoblastoma, 274.2.2 p53, 274.2.3 Adenomatous Polyposis Coli, 294.2.4 Phosphatase and Tensin Homologue,

304.2.5 Transforming Growth Factor-�, 304.2.6 Heritable Cancer Syndromes, 32

5 Interventions, 325.1 Prevention Strategies, 325.2 Targets, 33

5.2.1 Biochemical Targets, 335.2.2 Cyclooxygenase-2 and Cancer, 335.2.3 Other Targets, 35

5.3 Therapy, 355.3.1 Importance of Studying Gene

Expression, 355.3.2 cDNA Microarray Technology, 355.3.3 Discoveries from cDNA Microarray

Data, 37

5.3.4 Limitations of MicroarrayTechnologies, 37

5.4 Modifying Cell Adhesion, 375.4.1 MMP Inhibitors, 375.4.2 Anticoagulants, 385.4.3 Inhibitors of Angiogenesis, 38

5.5 Prospects for Gene Therapy of Cancer, 395.5.1 Gene Delivery Systems, 39

5.5.1.1 Viral Vectors, 405.5.1.2 Non-Viral Gene Delivery

Systems, 425.6 Gene Therapy Approaches, 43

5.6.1 Immunomodulation, 435.6.2 Suicidal Gene Approach, 445.6.3 Targeting Loss of Tumor Suppressor

Function and OncogeneOverexpression, 44

5.6.4 Angiogenesis Control, 455.6.5 Matrix Metalloproteinase, 45

6 Acknowledgments, 46

1 INTRODUCTION

Cancer is a major human health problemworldwide and is the second leading cause ofdeath in the United States (1). Over the past30 years, significant progress has beenachieved in understanding the molecular basisof cancer. The accumulation of this basicknowledge has established that cancer is a va-riety of distinct diseases and that defectivegenes cause these diseases. Further, gene de-fects are diverse in nature and can involve ei-ther loss or gain of gene functions. A numberof inherited syndromes associated with in-creased risk of cancer have been identified.

This chapter will review our current under-standing of the mechanisms of cancer develop-ment, or carcinogenesis, and the genetic basisof cancer. The roles of gene defects in bothgermline and somatic cells will be discussed asthey relate to genetic and sporadic forms ofcancer. Specific examples of oncogenes, or can-cer-causing genes, and tumor suppressorgenes will be presented, along with descrip-tions of the relevant pathways that signal nor-mal and cancer phenotypes.

While cancer is clearly associated with anincrease in cell number, alterations in mecha-nisms regulating new cell birth, or cell prolif-eration, are only one facet of the mechanismsof cancer. Decreased rates of cell death, or ap-

optosis, are now known to contribute to cer-tain types of cancer. Cancer is distinctive fromother tumor-forming processes because of itsability to invade surrounding tissues. Thischapter will address mechanisms regulatingthe important cancer phenotypes of alteredcell proliferation, apoptosis, and invasiveness.

Recently, it has become possible to exploitthis basic information to develop mechanism-based strategies for cancer prevention andtreatment. The success of both public and pri-vate efforts to sequence genomes, includinghuman and other organisms, has contributedto this effort. Several examples of mechanism-based anti-cancer strategies will be discussed.Finally, potential strategies for gene therapyof cancer will also be addressed.

2 TUMORIGENESIS

2.1 Normal-Precancer-Cancer Sequence

Insight into tumor development first camefrom epidemiological studies that examinedthe relationship between age and cancer inci-dence that showed that cancer incidence in-creases with roughly the fifth power of elapsedage (2). Hence, it was predicted that at leastfive rate-limiting steps must be overcome be-fore a clinically observable tumor could arise.It is now known that these rate-limiting steps

Molecular Biology of Cancer2

Page 3: Molecular Biology of Cancer - UC · PDF fileDepartments of Radiation Oncology/Cancer Biology Section Molecular and Cellular Biology ... 2 Molecular Biology of Cancer. are genetic mutations

are genetic mutations that dysregulate the ac-tivities of genes that control cell growth, reg-ulate sensitivity to programmed cell death,and maintain genetic stability. Hence, tumor-igenesis is a multistep process.

Although the processes that occur duringtumorigenesis are only incompletely under-stood, it is clear that the successive accumula-tion of mutations in key genes is the force thatdrives tumorigenesis. Each successive muta-tion is thought to provide the developing tu-mor cell with important growth advantagesthat allow cell clones to outgrow their morenormal neighboring cells. Hence, tumor devel-opment can be thought of as Darwinian evolu-tion on a microscopic scale with each succes-sive generation of tumor cell more adapted toovercoming the social rules that regulate thegrowth of normal cells. This is called clonalevolution (3).

Given that tumorigenesis is the result ofmutations in a select set of genes, much effortby cancer biologists has been focused on iden-tifying these genes and understanding howthey function to alter cell growth. Early effortsin this area were lead by virologists studyingretrovirus-induced tumors in animal models.These studies led to cloning of the first onco-genes and the realization that oncogenes, in-deed all cancer-related genes, are aberrantforms of genes that have important functionsin regulating normal cell growth (4). In subse-quent studies, these newly identified onco-genes were introduced into normal cells in aneffort to reproduce tumorigenesis in vitro. Im-portantly, it was found that no single onco-gene could confer all of the physiological traitsof a transformed cell to a normal cell. Ratherthis required that at least two oncogenes act-ing cooperatively to give rise to cells with thefully transformed phenotype (5). This obser-vation provides important insights into tu-morigenesis. First, the multistep nature of tu-morigenesis can be rationalized as mutationsin different genes with each event providing aselective growth advantage. Second, oncogenecooperativity is likely to be cause by the re-quirement for dysregulation of cell growth atmultiple levels.

Fearon and Vogelstein (6) have proposed alinear progression model (Fig. 1.1) to describetumorigenesis using colon carcinogenesis in

humans as the paradigm. They suggest thatmalignant colorectal tumors (carcinomas)evolve from preexisting benign tumors (ade-nomas) in a stepwise fashion with benign, lessaggressive lesions giving rise to more lethalneoplasms. In their model, both genetic [e.g.,adenomatous polyposis coli (APC) mutations]and epigenetic changes (e.g., DNA methyl-ation affecting gene expression) accumulateover time, and it is the progressive accumula-tion of these changes that occur in a preferred,but not invariable, order that are associatedwith the evolution of colonic neoplasms. Otherimportant features of this model are that atleast four to five mutations are required forthe formation of a malignant tumor, in agree-ment with the epidemiological data, withfewer changes giving rise to intermediate be-nign lesions, that tumors arise through themutational activation of oncogenes and inac-tivation of tumor suppressor genes, and that itis the sum total of the effect of these mutationson tumor cell physiology that is importantrather than the order in which they occur.

An important implication of the multistepmodel of tumorigenesis is that lethal neo-plasms are preceded by less aggressive inter-mediate steps with predictable genetic alter-ations. This suggests that if the genetic defectswhich occur early in the process can be identi-fied, a strategy that interferes with theirfunction might prevent development of moreadvanced tumors. Moreover, preventive screen-ing methods that can detect cells with theearly genetic mutations may help to identifythese lesions in their earliest and most curablestages. Consequently, identification of thegenes that are mutated in cancers and eluci-dation of their mechanism of action is impor-tant not only to explain the characteristic phe-notypes exhibited by tumor cells, but also toprovide targets for development of therapeu-tic agents.

2.2 Carcinogenesis

Carcinogenesis is the process that leads to ge-netic mutations induced by physical or chem-ical agents. Conceptually, this process can bedivided into three distinct stages: initiation,promotion, and progression (7). Initiation in-volves an irreversible genetic change, usuallya mutation in a single gene. Promotion is gen-

2 Tumorigenesis 3

Page 4: Molecular Biology of Cancer - UC · PDF fileDepartments of Radiation Oncology/Cancer Biology Section Molecular and Cellular Biology ... 2 Molecular Biology of Cancer. are genetic mutations

erally associated with increased proliferationof initiated cells, which increases the popula-tion of initiated cells. Progression is the accu-mulation of more genetic mutations that leadto the acquisition of the malignant or invasivephenotype.

In the best-characterized model of chemicalcarcinogenesis, the mouse skin model, initia-tion is an irreversible event that occurs when agenotoxic chemical, or its reactive metabolite,causes a DNA mutation in a critical growthcontrolling gene such as Ha-ras (8). Out-wardly, initiated cells seem normal. However,they remain susceptible to promotion and fur-ther neoplastic development indefinitely.DNA mutations that occur in initiated cellscan confer growth advantages, which allowthem to evolve and/or grow faster bypassingnormal cellular growth controls. The differenttypes of mutations that can occur includepoint mutations, deletions, insertions, chro-mosomal translocations, and amplifications.Three important steps involved in initiationare carcinogen metabolism, DNA repair, andcell proliferation. Many chemical agents mustbe metabolically activated before they becomecarcinogenic. Most carcinogens, or their activemetabolites, are strong electrophiles and bindto DNA to form adducts that must be removedby DNA repair mechanisms (9). Hence, DNArepair is essential to reverse adduct formationand to prevent DNA damage. Failure to repairchemical adducts, followed by cell prolifera-tion, results in permanent alterations or mu-tation(s) in the genome that can lead to onco-gene activation or inactivation of tumorsuppressor genes.

Promotion is a reversible process in whichchemical agents stimulate proliferation of ini-tiated cells. Typically, promoting agents arenongenotoxic, that is they are unable to formDNA adducts or cause DNA damage but areable to stimulate cell proliferation. Hence, ex-posure to tumor promoting agents results inrapid growth of the initiated cells and theeventual formation of non-invasive tumors. Inthe mouse skin tumorigenesis model, applica-tion of a single dose of an initiating agent doesnot usually result in tumor formation. How-ever, when the initiation step is followed byrepeated applications of a tumor promotingagent, such as 12-O-tetradecanoyl-phorbol-13-acetate (TPA), numerous skin tumors ariseand eventually result in invasive carcinomas.Consequently, tumor promoters are thoughtto function by fostering clonal selection of cellswith a more malignant phenotype. Impor-tantly, tumor formation is dependent on re-peated exposure to the tumor promoter. Halt-ing application of the tumor promoterprevents or reduces the frequency with whichtumors form. The sequence of exposure is im-portant because tumors do not develop in theabsence of an initiating agent even if the tu-mor promoting agent is applied repeatedly.Therefore, the genetic mutation caused by theinitiating agent is essential for further neo-plastic development under the influence of thepromoting agent.

Progression refers to the process of acquir-ing additional mutations that lead to malig-nancy and metastasis. Many initiating agentscan also lead to tumor progression, strong sup-port for the notion that further mutations are

Normalcoloncell

Hyper- proliferation

Earlyadenoma

Intermediateadenoma

Lateadenoma

Carcinoma Metastasis

Mutation ofAPC

DNAhypomethylation

Mutation ofK-ras Loss of DCC Loss of p53

Other geneticalterations

Figure 1.1. Adenoma-carcinoma sequence. Fearon and Vogelstein (6) proposed this classic modelfor the multistage progression of colorectal cancer. A mutation in the APC tumor suppressor gene isgenerally considered to be the initiation event. This is followed by the sequential accumulation ofother epigenetic and genetic changes that eventually result in the progression from a normal cell toa metastatic tumor.

Molecular Biology of Cancer4

Page 5: Molecular Biology of Cancer - UC · PDF fileDepartments of Radiation Oncology/Cancer Biology Section Molecular and Cellular Biology ... 2 Molecular Biology of Cancer. are genetic mutations

needed for cells to acquire the phenotypiccharacteristics of malignant tumor cells. Someof these agents include benzo(a)pyrene,�-napthylamine, 2-acetylaminofluorene,aflatoxin B1, dimethylnitrosamine, 2-amino-3-methylimidazo(4,5-f)quinoline (IQ), benzi-dine, vinyl chloride, and 4-(methylnitros-amino)-1-(3-pyridyl)-1-butanone (NNK) (10).These chemicals are converted into positivelycharged metabolites that bind to negativelycharged groups on molecules like proteins andnucleic acids. This results in the formation ofDNA adducts which, if not repaired, lead tomutations (9) (Fig. 1.2). The result of thesemutations enables the tumors to grow, invadesurrounding tissue, and metastasize.

Damage to DNA and the genetic mutationsthat can result from them are a central themein carcinogenesis. Hence, the environmentalfactors that cause DNA damage are of greatinterest. Environmental agents that can causeDNA damage include ionizing radiation, ultra-violet (UV) light, and chemical agents (11).Some of the DNA lesions that can result in-clude single-strand breaks, double-strandbreaks, base alterations, cross-links, insertionof incorrect bases, and addition/deletion ofDNA sequences. Cells have evolved severaldifferent repair mechanisms that can reversethe lesions caused by these agents, which hasbeen extensively reviewed elsewhere (12).

The metabolic processing of environmentalcarcinogens is also of key importance becausethis can determine the extent and duration towhich an organism is exposed to a carcinogen.Phase I and phase II metabolizing enzymes

play important roles in the metabolic activa-tion and detoxification of carcinogenic agents.The phase I enzymes include monooxygen-ases, dehydrogenases, esterases, reductases,and oxidases. These enzymes introduce func-tional groups on the substrate. The most im-portant superfamily of the phase I enzymesare the cytochrome P450 monooxygenases,which metabolize polyaromatic hydrocarbons,aromatic amines, heterocyclic amines, and ni-trosamines. Phase II metabolizing enzymesare important for the detoxification and excre-tion of carcinogens. Some examples includeepoxide hydrase, glutathione-S-transferase,and uridine 5�-diphosphate (UDP) glucuro-nide transferase. There are also some directacting carcinogens that do not require meta-bolic activation. These include nitrogen mus-tard, dimethylcarbamyl chloride, and �-pro-piolactone.

2.3 Genetic Variability and Other Modifiersof Tumorigenesis

2.3.1 Genetic Variability Affecting Cancer.Different types of cancers, as well as their se-verity, seem to correlate with the type of mu-tation acquired by a specific gene. Mutation“hot spots” are regions of genes that are fre-quently mutated compared with other regionswithin that gene. For example, observationsthat the majority of colon adenomas are asso-ciated with alterations in the adenomatouspolyposis coli (APC) have been based on im-munohistochemical analysis of �-catenin lo-calization and formation of less than full

Procarcinogen CarcinogenMetabolic activation

Formation ofcarcinogen-DNA adduct

DNA binding

DNA repair

Normal cellDNAreplication

Initiated cell

Celldeath

Detoxification

Excretion ofmetabolites

Figure 1.2. Possible outcomes ofcarcinogen metabolic activation.Once a carcinogen is metabolicallyactivated it can bind to DNA andform carcinogen-DNA adducts. Theseadducts will ultimately lead to muta-tions if they are not repaired. If DNArepair does not occur, the cell will ei-ther undergo apoptosis or the DNAwill be replicated, resulting in an ini-tiated cell.

2 Tumorigenesis 5

Page 6: Molecular Biology of Cancer - UC · PDF fileDepartments of Radiation Oncology/Cancer Biology Section Molecular and Cellular Biology ... 2 Molecular Biology of Cancer. are genetic mutations

length APC protein production after in vitrotranslation of colonic mucosal tissue RNA.These studies have not documented specificgene mutations in APC. This is important, be-cause it is known from animal studies that thelocation of APC mutations can have a dra-matic effect on the degree of intestinal carci-nogenesis. Thus, it is possible that colon ade-noma size, and subsequent risk of colon cancercould be dictated by location of specific muta-tions in APC (Fig. 1.3).

As suggested by the model depicted in Fig.1.3, high risk might be associated with muta-tions causing stop codons in the amino termi-nal end of the protein. Low risk might be as-sociated with mutations resulting in peptidesof greater length. Current research is testingthe hypothesis that specific genetic alterationsin APC alone may be sufficient as a prognosticfactor for risk of adenoma recurrence and sub-sequently, colon cancer development.

One type of genetic alteration that is gain-ing increasing attention is the single nucleo-tide polymorphism (SNP). This polymorphism

results from a single base mutation that leadsto the substitution of one base for another.SNPs occur quite frequently (about every 0.3–1 kb within the genome) and can be identifiedby several different techniques. A commonmethod for the analysis of SNPs is based onthe knowledge that single-base changes havethe capability of destroying or creating a re-striction enzyme site within a specific regionof DNA. Digestion of a piece of DNA, contain-ing the site in question, with the appropriateenzyme can distinguish between variantsbased on the resulting fragment sizes. Thistype of analysis is commonly referred to asrestriction fragment length polymorphism(RFLP).

The importance of analyzing SNPs rests onthe premise that individuals with a nucleotideat a specific position may display a normalphenotype, whereas individuals with a differ-ent nucleotide at this same position may ex-hibit increased predisposition for a certain dis-ease or phenotype. Therefore, many studiesare being conducted to determine the fre-

0 2843

APC∆716 Min APC∆1638 Min

Armadillorepeats

453 −766

Mutationclusterregion

DrosophiliaDLG binding2771−2843

Homodimerizationregion1−71

Microtubulebinding

2143−2843

EB1 binding2143−2843

Murine models Intestinal tumor number

APC∆716 200−600Min (850 stopcodon) 60−80

APC∆1638 <10

Figure 1.3. Diagram of APC protein regions, relating risk of intestinal carcinogenesis to length ofAPC peptide translated. APC contains 2833 amino acids. Mutation hot spot regions are found in areasbetween amino acids 1500–2000. Three genetically altered mouse models of APC-dependent intes-tinal carcinogenesis have been developed. Min mice have a stop codon mutation in codon 850 of themurine APC homolog. Two transgenic mice, APC�716 and APC�1635, also have been developed.Intestinal tumor number in these models is inversely related to size of the APC peptide translated.

Molecular Biology of Cancer6

Page 7: Molecular Biology of Cancer - UC · PDF fileDepartments of Radiation Oncology/Cancer Biology Section Molecular and Cellular Biology ... 2 Molecular Biology of Cancer. are genetic mutations

quency of specific SNPs in the general popula-tion and to use these findings to explain phe-notypic variation.

For example, a recent study found an asso-ciation between a polymorphism leading to anamino acid substitution (aspartate to valine)in codon 1822 of the APC gene and a reducedrisk for cancer in people eating a low-fat diet(13). The variant valine had an allele fre-quency of 22.8% in a primarily Caucasian con-trol population. This non-truncating muta-tion has not yet been shown to have functionalsignificance. If functional, such a polymor-phism could cooperate with single allele trun-cating mutations that occur with high fre-quency in sporadic colon adenomas (14), toincrease colon cancer risk. This polymorphismis especially interesting, because dietary fac-tors, specifically fat consumption, may con-tribute to risk in only specific genetic subsets.

2.3.2 Genetic Variability in c-myc–Depen-dent Expression of Ornithine Decarboxylase.The proliferation-associated polyamines areessential for cell growth but may contribute tocarcinogenesis when in excess. Various stud-ies have shown that inhibition of polyaminesynthesis impedes carcinogenesis. Ornithinedecarboxylase (ODC), the first enzyme in poly-

amine synthesis, may play a key role in tumordevelopment. Therefore, elucidation of themechanisms by which ODC is regulated is es-sential. The literature indicates that ODC is adownstream mediator of APC and suggeststhat ODC may be an APC modifier gene. Thus,polymorphisms in the ODC promoter affect-ing c-myc–dependent ODC transcriptioncould be a mechanism of genetic variability ofAPC-dependent carcinogenesis.

O’Brien and colleagues (15) have measuredthe incidence in several human subgroups of aSNP in a region of the ODC promoter, 3� of thetranscription start site, that is flanked by twoE-boxes (CACGTG) (Fig. 1.4). The E-box is aDNA sequence where specific transcriptionfactors bind. The two resulting alleles areidentified by a polymorphic PstI RFLP. Theminor allele (A at position �317) is homozy-gous in 6–10% of individuals, whereas the ma-jor allele (G at position �317) is homozygousor heterozygous in 90–94% of these groups.They have also measured functionality of thepolymorphisms. When ODC promoter-re-porter constructs are expressed in rodentcells, the minor allele confers 3–8 times thepromoter activity compared with the major al-lele. Further, expression of the minor allele is

ODC gene

e-box (1) e-box (2) e-box (3)

+300

G/A SNP

E-box (1) SNP allele (Frequency) Promoter activity

CACGTG G (90−95%) 1CAGCTG G (90−95%) 0.5CACGTG A (5−10%) 3−8

Figure 1.4. Influence of specific genetic changes on ODC promoter activity. These data were derivedfrom transient transfection experiments in human colon tumor–derived HT29 cells. The arrow inthis figure 1.4 shows the SNP. The SNP occurs between two E-boxes that are located 3� of thetranscription start site. The effects of this genetic change are taken from Guo et al. (56). It isimportant to point out that the constructs used to assess the promoter activity of the polymorphicregion containing the SNP and E-boxes 2 and 3 contained some of the 5� promoter region, but notE-box 1 (56). The constructs used to assess the role of E-box 1 in HT-29 contained the major, c-mycunresponsive allele between E-boxes 2 and 3.

2 Tumorigenesis 7

Page 8: Molecular Biology of Cancer - UC · PDF fileDepartments of Radiation Oncology/Cancer Biology Section Molecular and Cellular Biology ... 2 Molecular Biology of Cancer. are genetic mutations

enhanced by c-myc expression to a greater ex-tent than the major allele.

2.4 Epigenetic Changes

Gene function can be disrupted either throughgenetic alterations, which directly mutate ordelete genes, or epigenetic alterations, whichalter the state of gene expression. Epigeneticmechanisms regulating gene expression in-clude signal transduction pathways, DNAmethylation, and chromatin remodeling.Methylation of DNA is a biochemical additionof a methyl group at position 5 of the pyrimi-dine ring of cytosine in the sequence CG. Thismodification occurs in two ways: (1) from apreexisting pattern on the coding strand or (2)by de novo addition of a methyl group to fullyunmethylated DNA. Cleavage of DNA withthe restriction endonuclease HpaII, whichcannot cut the central C in the sequenceCCGG if it is methylated, allows detection ofmethylated sites in DNA. Small regions ofDNA with methylated cytosine, called “CpGislands,” have been found in the 5�-promoterregion of about one-half of all human genes(including most housekeeping genes).

There are three DNA methyltransferases(Dnmt), Dnmt1, Dnmt3a, and Dnmt3b, thathave been identified in mammalian cells (16).The most abundant and ubiquitous enzyme,Dnmt1, shows high affinity for hemimethyl-ated DNA, suggesting a role of Dnmt1 in theinheritance of preexisting patterns of DNAmethylation after each round of DNA replica-tion. The other two enzymes, Dnmt3a andDnmt3b, are tissue specific and have beenshown to be involved in de novo methylation.De novo CpG island methylation, however, isnot a feature of proliferating cells, and can beconsidered a pathologic event in neoplasia.

Over the years, a number of differentmethyl-CpG binding proteins, such as methyl-CpG-binding domain-containing proteins(MBD1-4) were identified (17) that competewith transcription factors and prevent themfrom binding to promoter sequences. Thesemethyl-CpG binding factors can also recruithistone deacetylases (HDACs), resulting incondensation of local chromatin structure(Fig. 1.5). This makes the methylated DNAless accessible to transcription factors and re-sults in gene silencing.

Gene expression is inhibited by DNA meth-ylation. DNA methylation patterns dramati-cally change at different stages of cell develop-ment and differentiation and correlate withchanges in gene expression (18). Demethyl-ation releases gene expression in the first daysof embryogenesis. Later, de novo methylationestablishes adult patterns of gene methyl-ation. In differentiated cells, methylation sta-tus is retained by the activity of the Dnmt1enzyme. In normal tissues, DNA methylationis associated with gene silencing, chromosomeX inactivation (19), and imprinting (20). Be-cause the most normal methylation takesplace within highly repeated transposable ele-ments, it has been proposed that such methyl-ation plays a role in genome defense by sup-pressing potentially harmful effects ofexpression at these sites.

Neoplastic cells are characterized by simul-taneous global DNA hypomethylation, local-ized hypermethylation that involves CpG is-lands and increased HDAC activity (21).Hypomethylation has been linked to chromo-somal instability in vitro and it seems to havethe same effect in carcinogenesis (22). 5-Meth-ylcytosine is a relatively unstable base becauseits spontaneous deamination leads to the for-mation of uracil. Such changes can also con-tribute to the appearance of germline muta-tions in inherited disease and somaticmutations in neoplasia. Aberrant CpG islandhypermethylation in normally unmethylatedregions around gene transcription start sites,which results in transcriptional silencing ofgenes, suggests that it plays an important roleas an alternate mechanism by which tumorsuppressor genes are inactivated in cancer(21). Hypermethylated genes identified in hu-man cancers include the tumor suppressorgenes that cause familial forms of human can-cer when mutated in the germline, as well asgenes that are not fully documented tumorsuppressors (Table 1.1). Some of these genes,such as APC, the breast cancer gene BRCA-1,E-cadherin, mismatch repair gene hMLH1,and the Von Hippel-Lindau gene can exhibitthis change in non-familial cancers.

Recent studies indicate that promoter hy-permethylation is often an early event in tu-mor progression. It has been shown in thecolon that genes that have increased hyper-

Molecular Biology of Cancer8

Page 9: Molecular Biology of Cancer - UC · PDF fileDepartments of Radiation Oncology/Cancer Biology Section Molecular and Cellular Biology ... 2 Molecular Biology of Cancer. are genetic mutations

methylation in the promoter region in normaltissue as a function of aging are the same asgenes with the highest rate of promoter hyper-methylation in tumors (9). Interestingly, thisgroup of genes does not include classic tumorsuppressor genes. Some genes, such as the es-trogen receptor where age-related hypermeth-ylation in the colon was first discovered, maybe important for the modulation of cell growthand differentiation in the colonic mucosa.

Promoter hypermethylation of genes,which are normally unmethylated at all ages,has also been found early in tumorigenesis.These epigenetic alterations can produce theearly loss of cell cycle control, altered regula-tion of gene transcription factors, disruptionof cell-cell interactions, and multiple types ofgenetic instability, which are all characteristicof neoplasia. For example, hypermethylationof the APC gene has recently been reported fora subset of colon cancers (23). Hypermethyl-ation of hMLH1, which is associated with mi-crosatellite instability in colon, endometrial,

and gastric neoplasia, has been seen in earlystages of cancer progression (24). Finally, hy-permethylation of the E-cadherin promoterfrequently occurs in early stages of breast can-cer and can trigger invasion (25).

Loss of gene function through epigeneticchanges differs from genetic changes in termsof its consequences for tumor biology. First,gene function loss caused by aberrant pro-moter methylation may manifest in a moresubtle, selective advantage than gene muta-tions during tumor progression. Second, al-though promoter hypermethylation causinggene silencing is usually stable in cancer cells,this change, unlike mutation, is potentially re-versible. It has become evident that not onlythe mutagens, but various factors influencingcell metabolism, particularly methylation, lieat the origin of carcinogenesis.

Silencing of gene expression by methyl-ation may be modulated by biochemical or bi-ological manipulation. It has been shown thatpharmacological inhibition of methyltrans-

HDAC HDAC

Active gene

De novomethylation

Recruitment of MBPand HDAC

Condensed chromatin-silenced gene

Deacetylated histones

Inactive gene

Active (?) gene

Figure 1.5. Effect of methylation and histone deacetylation on gene expression. When a gene isactive, the promoter region is occupied by transcription factors that direct production of messengerRNA. De novo methylation has minimal effects on gene expression. However, methylated DNAattracts methyl-binding proteins (MBP). These methyl-binding proteins in turn attract a proteincomplex that contains histone deacetylase (HDAC). This results in inhibition of messenger RNAsynthesis, and no functional protein can be made from the gene. Through the action of MBP andHDAC, the DNA structure changes to a compact, “condensed chromatin” configuration, which re-sults in permanent inhibition of messenger RNA and protein synthesis (silencing).

2 Tumorigenesis 9

Page 10: Molecular Biology of Cancer - UC · PDF fileDepartments of Radiation Oncology/Cancer Biology Section Molecular and Cellular Biology ... 2 Molecular Biology of Cancer. are genetic mutations

ferases resulted in reactivation of gene expres-sion in vitro (26) and prevented tumor growthin animal models (27). These studies gener-ated interest in the clinical uses of hypomethy-lating agents in humans.

3 MOLECULAR BASIS OF CANCERPHENOTYPES

Cancer is a multistep process that requires theaccumulation of multiple genetic mutations ina single cell that bestow features characteris-tic of a neoplastic cell. Typically, tumor cellsdiffer from normal cells in that they exhibituncontrolled growth. Because features thatdistinguish tumor from normal cells may bekey to understanding neoplastic cell behaviorand may ultimately lead to therapies that cantarget tumor cells, considerable effort hasbeen directed at identifying the phenotypiccharacteristics of in vitro–transformed cellsand of tumor cells derived from naturalsources. This work has resulted in a list of

properties that are characteristic of tumorcells and that are now known to be the basisfor the behaviors exhibited by neoplastic cells.Some of the features that will be discussed indetail include immortality, decreased depen-dence on growth factors to support prolifera-tion, loss of anchorage-dependent growth, lossof cell cycle control, reduced sensitivity to ap-optotic cell death, and increased genetic insta-bility. Other morphological and biochemicalcharacteristics used to identify the trans-formed phenotype are cytological changes, al-tered enzyme production, and the ability toproduce tumors in experimental animals (28).

3.1 Immortality

Normal diploid fibroblasts have a limited ca-pacity to grow and divide both in vivo and invitro. Even if provided with optimal growthconditions, in vitro normal cells will cease di-viding after 50–60 population doublings andthen senesce and die. In contrast, malignantcells that have become established in culture

Table 1.1 Hypermethylated Genes in Cancer

Gene Function Type of Tumor

Familial CancersAPC Signal transduction Colon cancerBRCA1 DNA repair Breast cancerE-cadherin Adhesion and metastasis Multiple cancershMLH1 DNA mismatch repair Colon, gastric, and endometrial

cancerp16/CDKN2A Cell cycle regulation Multiple cancersRB1 Cell cycle regulation RetinoblastomaVHL Cytoskeletal organization, angiogenesis

inhibitionRenal-cell cancer

Other CancersAndrogen receptor Growth and differentiation Prostate cancerc-ABL Tyrosine kinase Chronic myelogenous leukemiaEndothelin receptor B Growth and differentiation Prostate cancerEstrogen receptor � Transcription Multiple cancersFHIT Detoxification Esophageal cancerGST-� Drug transport Prostate cancerMDR1 Drug transport Acute leukemiasO6-MGMT DNA repair Multiple cancersp14/ARF Cell cycle regulation Colon cancerp15/CDKN2B Cell cycle regulation Malignant hematologic diseaseProgesterone receptor Growth and differentiation Breast cancerRetinoic acid receptor � Growth and differentiation Colon and breast cancerTHBS1 Angiogenesis inhibition Colon cancer, glioblastoma

multiformeTIMP3 Metastasis Multiple cancers

Molecular Biology of Cancer10

Page 11: Molecular Biology of Cancer - UC · PDF fileDepartments of Radiation Oncology/Cancer Biology Section Molecular and Cellular Biology ... 2 Molecular Biology of Cancer. are genetic mutations

proliferate indefinitely and are said to be im-mortalized. The barrier that restricts the lifespan of normal cells is known as the Hayflicklimit and was first described in experimentsthat attempted immortalization of rodentcells (29). Normal embryo-derived rodentcells, when cultured in vitro, initially dividerapidly. Eventually, however, these culturesundergo a crisis phase during which many ofthe cells senesce and die. After extended main-tenance, however, proliferation in the culturesincreases and cells that can divide indefinitelyemerge. The molecular changes that takeplace during crisis have revealed at least twoimportant restrictions that must be overcomefor cells to become immortalized and both ofthese changes occur in natural tumor cells.

One barrier to cellular immortalization isthe inability of the DNA replication machin-ery to efficiently replicate the linear ends ofDNA at the 5� ends, which leads to the short-ening of the chromosome. In bacteria, the end-replication problem is solved with a circularchromosome. In human cells, the ends of chro-mosomes are capped with 5–15 kb of repetitiveDNA sequences known as telomeres. Telo-meres serve as a safety cap of noncoding DNAthat is lost during normal cell division withoutconsequence to normal function of the cell.However, because telomere length is short-ened with each round of cell division, indefi-nite proliferation is impossible because even-tually the inability to replicate chromosomalends nibbles into DNA containing vital genes.

Telomeres seem to be lengthened duringgametogenesis as a consequence of the activityof an enzyme called telomerase. Telomeraseactivity has been detected in normal ovarianepithelial tissue. More importantly, telomer-ase activity is elevated in the tumor tissue butnot the normal tissue from the same patient.This implies that one mechanism by which tu-mor cells overcome the shortening telomereproblem and acquire the capacity to prolifer-ate indefinitely is through abnormal up-regu-lation of telomerase activity. The finding thattelomerase activity is found almost exclusivelyin tumor cells is significant because it suggeststhat this enzyme may be a useful therapeutictarget (30). Therapies aimed at suppressingtelomerase would eliminate a feature essentialfor tumor cell survival and would be selective.

A second feature of immortalization is lossof growth control by elimination of tumor sup-pressor activity. Recent evidence suggeststhat inactivating mutations in both the Rband p53 tumor suppressor genes occurs dur-ing crisis. Both of these genes are discussed inmore detail later in this chapter and both func-tion to inhibit cell proliferation by regulatingcell cycle progression. Consequently, loss oftumor suppressor function also appears to be acritical event in immortalization.

3.2 Decreased Dependence on GrowthFactors to Support Proliferation

Cells grown in culture require media supple-mented with various growth factors to con-tinue proliferating. In normal human tissues,growth factors are generally produced extra-cellularly at distant sites and then are eithercarried through the bloodstream or diffuse totheir nearby target cells. The former mode ofgrowth factor stimulation is termed endocrinestimulation, and the latter mode, paracrinestimulation. However, tumor cells often pro-duce their own growth factors that bind to andstimulate the activity of receptors that are alsopresent on the same tumor cells that are pro-ducing the growth factor. This results in a con-tinuous self-generated proliferative signalknown as autocrine stimulation that drivesproliferation of the tumor cell continuouslyeven in the absence of any exogenous prolifer-ative signal. Autocrine stimulation is mani-fested as a reduced requirement for serum be-cause serum is the source of many of thegrowth factors in the media used to propagatecells in vitro.

Because of the prominent role that growthfactors and their cognate receptors play in tu-mor cell proliferation, they have also becomefavorite therapeutic targets. For example, theepidermal growth factor receptor (EGFR) isknown to play a major role in the progressionof most human epithelial tumors, and its over-expression is associated with poor prognosis.As a consequence, different approaches havebeen developed to block EGFR activationfunction in cancer cells, including anti-EGFRblocking monoclonal antibodies (MAb), epi-dermal growth factor (EGF) fused to toxins,and small molecules that inhibit the receptor’styrosine kinase activity (RTK). Of these, an

3 Molecular Basis of Cancer Phenotypes 11

Page 12: Molecular Biology of Cancer - UC · PDF fileDepartments of Radiation Oncology/Cancer Biology Section Molecular and Cellular Biology ... 2 Molecular Biology of Cancer. are genetic mutations

orally active anilinoquinazoline, ZD1839(“Iressa”) shows the most promise as an anti-tumor agent by potentiating the antitumor ac-tivity of conventional chemotherapy (31).

3.3 Loss of Anchorage-Dependent Growthand Altered Cell Adhesion

Most normal mammalian cells do not grow,but instead undergo cell death if they becomedetached from a solid substrate. Tumor cells,however, frequently can grow in suspension orin a semisolid agar gel. The significance of theloss of this anchorage-dependent growth ofcancer cells relates to the ability of the parenttumor cells to leave the primary tumor siteand become established elsewhere in the body.The ability of cancer cells to invade and metas-tasize foreign tissues represents the final andmost difficult-to-treat stage of tumor develop-ment, and it is this change that accompaniesthe conversion of a benign tumor to a life-threatening cancer.

Metastasis is a complex process that re-quires the acquisition of several new charac-teristics for tumor cells to successfully colo-nize distant sites in the body. Epithelial cellsnormally grow attached to a basement mem-brane that forms a boundary between the ep-ithelial cell layers and the underlying support-ing stroma separating the two tissues. Thisbasement membrane consists of a complex ar-ray of extracellular matrix proteins includingtype IV collagen, proteoglycans, laminin, andfibronectin, which normally acts as a barrierto epithelial cells. A common feature of tumorcells with metastatic potential is the capacityto penetrate the basement membrane by pro-teolysis, to survive in the absence of attach-ment to this substrate, and to colonize andgrow in a tissue that may be foreign relative tothe original tissue of origin.

Consequently, metastasis is a multistepprocess that begins with detachment of tumorcells from the primary tumor and penetrationthrough the basement membrane by degrada-tion of the extracellular matrix (ECM) pro-teins. This capacity to proteolytically degradebasement membrane proteins is driven, inpart, by the expression of matrix metallo-proteinases. Matrix metalloproteinases, orMMPs, are a family of enzymes that are eithersecreted (MMPs 1–13, 18–20) or anchored in

the cell membrane (MMPs 14–17) (Table 1.2).Regulation of MMPs occurs at several levels:transcription, proteolytic activation of the zy-mogen, and inhibition of the active enzyme(32). MMPs are typically absent in normaladult cells, but a variety of stimuli, such ascytokines, growth factors, and alterations incell-cell and cell-ECM interactions, can inducetheir expression. The expression of MMPs intumors is frequently localized to stromal cellssurrounding malignant tumor cells. Most ofthe MMPs are secreted in their inactive (zy-mogen) form and require proteolytic cleavageto be activated. In some cases, MMPs havebeen shown to undergo mutual and/or autoac-tivation in vitro (33).

Several lines of evidence implicate MMPsin tumor progression and metastasis. First,MMPs are overexpressed in tumors from a va-riety of tissues and the expression of one, ma-trilysin, is clearly elevated in invasive prostatecancer epithelium (34–36). Second, reductionof tissue inhibitor of matrix metalloprotein-ases-1 (TIMP-1) expression in mouse fibro-blasts (Swiss 3T3), using antisense RNA tech-nology, increased the incidence of metastatictumors in immunocompromised mice. Simi-larly, overexpression of the various MMPs hasprovided direct evidence for their role in me-tastasis. Importantly, synthetic MMP inhibi-tors have also been produced and they lead toa reduction in metastasis in several experi-mental models of melanoma, colorectal carci-noma, and mammary carcinoma, suggesting amechanism by which the invasive potential oftumors may be reduced (37).

Once tumor cells escape through the base-ment membrane, they can metastasizethrough two major routes, the blood and lym-phatic vessels. Tumors originating in differentparts of the body have characteristic patternsof invasion. Some tumors, such as those of thehead and neck, spread initially to regionallymph nodes. Others, such as breast tumors,have the ability to spread to distant sites rela-tively early. The site of the primary tumorgenerally dictates whether the invasion willoccur through the lymphatic or blood vesselsystem. The cells that escape into the vascula-ture must evade host immune defense mecha-nisms to be successfully transported to re-gional or distal locations. Tumor cells then

Molecular Biology of Cancer12

Page 13: Molecular Biology of Cancer - UC · PDF fileDepartments of Radiation Oncology/Cancer Biology Section Molecular and Cellular Biology ... 2 Molecular Biology of Cancer. are genetic mutations

exit blood vessels and escape into the host tis-sue by again compromising a basement mem-brane, this time the basement membrane ofthe blood vessel endothelium. Projectionscalled invadopodia, which contain various pro-teases and adhesive molecules, adhere to thebasement membrane, and this involves mem-brane components such as laminin, fibronec-tin, type IV collagen, and proteoglycans. Thetumor cells then produce various proteolyticenzymes, including MMPs, which degrade thebasement membrane and allow invasion of thehost tissue. This process is referred to as ex-travasation.

The interaction between cells and extracel-lular matrix proteins occurs through cell-sur-face receptors, the best characterized of whichis the fibronectin receptor that binds fibronec-tin. Other receptors bind collagen and lami-nin. Collectively these receptors are called in-tegrins, and their interaction with matrixcomponents conveys regulatory signals to the

cell (38). They are heterodimeric moleculesconsisting of one of several alpha and beta sub-units that may combine in any number of per-mutations to generate a receptor with distinctsubstrate preferences. Changes in the expres-sion of integrin subunits is associated with in-vasive and metastatic cells facilitating inva-sion by shifting the cadre of integrins tointegrins that preferentially bind the de-graded subunits of extracellular matrix pro-teins produced by MMPs. Hence, integrin ex-pression has served as a marker for theinvasive phenotype and may be a logical targetfor novel therapies that interfere with theprogress of advanced tumors.

In addition to their role in invasion, theevidence also indicates that MMPs may play arole in tumor initiation and in tumorigenicity.Expression of MMP-3 in normal mammary ep-ithelial cells led to the formation of invasivetumors (39). A proposed mechanism for thisinitiation involves the ability of MMP-3 to

Table 1.2 MMPs

MMP Common Name Substrates Cell Surface

1 collagenase-1, interstitialcollagenase

collagen I, II, III, VII, X, IGFBP yes

2 gelatinase A gelatin, collagen I, IV, V, X, laminin, IGFBP,latent TGF-�

yes

3 stromelysin-1 collagen III, IV, V, IX, X, gelatin,E-cadherin, IGFBP, fibronectin,elastin, laminin proteoglycans, perlecan,HB-EGF, proMMP-13

unknown

7 matrilysin laminin, fibronectin, gelatin, collagen IV,proteoglycans FasL, proMMP-1, HB-EGF

yes

8 collagenase-2, neutrophilcollagenase

collagen I, II, III, VII, X unknown

9 gelatinase B collagen I, IV, V, X, gelatin, IGFBP, latentTGF-b

yes

10 stromelysin-2 collagen III, IV, IX, X, gelatin, laminin,proteoglycans, proMMP-1, proMMP-13

unknown

11 stromelysin-3 IGFBP, a-1-antiprotease unknown12 metalloelastase elastin, proMMP-13 unknown13 collagenase-3 collagen I, II, III, IV, VII, X, XIV,

fibronectin, proMMP-9, tenascin, aggrecanunknown

14 MT1-MMP gelatin, collagen I, fibrin, proteoglycans,laminin, fibronectin, proMMP-2

yes

15 MT2-MMP laminin, fibronectin, proMMP-2, proMMP-13, tenascin

yes

16 MT3-MMP gelatin, collagen III, fibronectin, proMMP-2 yes17 MT4-MMP unknown yes

18/19 RASI-1 unknown unknown20 Enamelysin amelogenin unknown

3 Molecular Basis of Cancer Phenotypes 13

Page 14: Molecular Biology of Cancer - UC · PDF fileDepartments of Radiation Oncology/Cancer Biology Section Molecular and Cellular Biology ... 2 Molecular Biology of Cancer. are genetic mutations

cleave E-cadherin. E-cadherin is a protein in-volved in cell-cell adhesion together withother proteins such as �-catenin and �-cati-nin. Loss of E-cadherin function is known tolead to tumorigenicity and invasiveness as aresult of loss of cellular adhesion. Interest-ingly, inhibition of MMP-7 and MMP-11, us-ing antisense approaches, did not affect inva-siveness or metastatic potential in vitro.However, tumorigenicity was altered (40).Matrilysin, MMP-7 messenger RNA (mRNA),are present in benign tumors and malignanttumor cells of the colon. The relative level ofmatrilysin expression correlates with thestage of tumor progression.

3.4 Cell Cycle and Loss of CellCycle Control

Proliferation is a complex process consistingof multiple subroutines that collectively bringabout cell division. At the heart of prolifera-tion is the cell cycle, which consists of manyprocesses that must be completed in a timelyand sequence specific manner. Accordingly,regulation of cell cycle events is a multifacetedaffair and consists of a series of checks andbalances that monitor nutritional status, cellsize, presence or absence of growth factors,and integrity of the genome. These cell cycleregulatory pathways and the signal transduc-tion pathways that communicate with themare populated with oncogenes and tumor sup-pressor genes.

Cell division is divided into four phases: G1,S, G2, and M (Fig. 1.6). The entire process ispunctuated by two spectacular events, thereplication of DNA during S phase and chro-mosome segregation during mitosis or Mphase. Of the four cell cycle phases, three canbe assigned to replicating cells and only the G1phase, and a related quiesent phase, G0, arenonreplicative in nature. Normal cycling cellsthat cease to proliferate enter the restingphase, or G1, and their exit into the replicativephases is strongly dependent on the presenceof growth factors and nutrients. However,once the cells enter the replicative phase of thecell cycle, they become irrevocably committedto completing cell division. Hence, the condi-tions that lead to exit from G1 and entry into Sare tightly regulated and are frequently mis-regulated in neoplastic cells that exhibit un-

controlled proliferation. Studies first con-ducted by Arthur Pardee revealed theexistence of a point in G1 that restricted thepassage of cells into S phase, and this was pos-tulated to be controlled by a labile protein fac-tor (41). Passage across this restriction point,or R point, is now known to be sensitive togrowth factor stimulation.

Movement through the cell cycle is con-trolled by two classes of cell cycle proteins,cyclins and cyclin dependent kinases (CDKs),which physically associate to form a proteinkinase that drives the cell cycle forward (42).At least 8 cyclins and 12 CDKs have been iden-tified in mammalian cells. The name “cyclin”derives from the characteristic rise and fall inabundance of cyclin B as cells progressthrough the cell cycle. The accumulation ofcyclin proteins occurs through cell cycle-de-pendent induction of gene transcription, butelimination of cyclins occurs by carefully reg-ulated degradation that is enabled throughprotein sequence tags known as destructionboxes and PEST sequences. Although not allof the cyclin types exhibit this oscillation inprotein quantity, those cyclins that play keyroles in progression through the cell cycle (cy-clins E, A, and B) are most abundant duringdiscrete phases of the cell cycle. Cyclin D1 issynthesized during G1 just before the restric-tion point and plays an important role in reg-ulation of the R point. Cyclin E is most abun-dant during late G1 and early S and isessential for exit from G1 and progression intoS phase. Elevated levels of these two G1 cyc-lins can result in uncontrolled proliferation.Indeed, both cyclin D1 and cyclin E are over-expressed in some tumor types, suggestingthat the cyclins and other components of thecell cycle may be useful therapeutic targets(43).

The second component of the enzyme com-plex is CDK that, as the name implies, re-quires an associated cyclin to become active.At least 12 of the protein kinases have beenisolated from humans, Xenopus, and Drosoph-ila, and are numbered according to a stan-dardized nomenclature beginning with CDK1,which for historical reasons, is most fre-quently referred to as cell division cycle 2(cdc2). Unlike the cyclins, abundance of theCDK proteins remains relatively constant

Molecular Biology of Cancer14

Page 15: Molecular Biology of Cancer - UC · PDF fileDepartments of Radiation Oncology/Cancer Biology Section Molecular and Cellular Biology ... 2 Molecular Biology of Cancer. are genetic mutations

throughout the cell cycle. Instead, their activ-ity changes during different phases of the cellcycle in accordance with whether or not anactivating cyclin is present and whether or notthe kinase itself is appropriately phosphory-lated. Both cyclins and CDKs are highly con-served from yeast to man and function simi-larly, suggesting that the cell cycle iscontrolled by a universal cell cycle engine thatoperates through the action of evolutionarilyconserved proteins. Hence, drug discoverystudies aimed at identifying agents that regu-late the cell cycle may be performed in modelorganisms, such as yeast, C. elegans, and Dro-sophila with some assurance that the targetedmechanisms will also be relevant to humans.

It is now clear that specific cyclin/cdk com-plexes are required during specific stages ofthe cell cycle. Cyclin D1/cdk4,6 activity is es-sential for crossing the restriction point andpushing cells into replication. A major sub-strate of the cyclin D1/cdk4,6 complex is theretinoblastoma (Rb) tumor suppressor pro-tein, which when phosphorylated by this ki-nase complex, is inactivated. This frees the cellfrom the restrictions on cell proliferation im-posed by the Rb protein. It is this event that isbelieved to be decisive in the stimulation ofresting cells to undergo proliferation. Cyclin

E/cdk2 plays a role later in the cell cycle forproliferating cells by pushing them from G1into S phase. Cyclin E is overexpressed insome breast cancers where it may enhance theproliferative capacity of tumor cells. CyclinA/cdk2 sustains DNA replication and is there-fore required during S phase. Cyclin B/cdc2 isrequired by cells entering mitosis up throughmetaphase. At the end of metaphase, cyclin Bis degraded, and cdc2 becomes inactivated, al-lowing mitotic cells to progress into anaphaseand to complete mitosis. Sustaining the activ-ity of cyclin B/cdc2 causes cells to arrest inmetaphase. Hence, it is the collective resultbrought about by the activation and deactiva-tion of cyclin/cdk complexes that pushes pro-liferating cells through the cell cycle.

Superimposed on the functions of the cellcycle engine is a complex network of both pos-itive and negative regulatory pathways. Im-portant negative regulators are the cyclin de-pendent kinase inhibitors or CKIs. There aretwo families of CKIs, the Cip/Kip family andthe INK4 family (44). The Cip/Kip family con-sists of three members, p21/Cip1/waf1/Sdi1,p21/Kip1, and p57/Kip2. All of the proteins inthis family have broad specificity and can bindto and inactivate most of the cyclin/cdk com-plexes that are essential for progression

Cyclin B

Cdk1

Cyclin A

Cdk2

Cyclin E

Cdk2

p53

p21

Cyclin D

Cdk4M

Rb

R

S

G2 G1

Cyclin D

Cdk6

Figure 1.6. Model of the cell cycle andthe cyclin/cdk complexes that are re-quired at each cell cycle phase. CyclinD/cdk4-6 complexes suppress Rb functionby phosphorylating the protein allowingtransition across the restriction R-point.P53 suppresses cell cycle progression bystimulating the expression of the cyclindependent kinase inhibitor p21, whichbinds with and inactivates a variety ofcyclin/cdk complexes.

3 Molecular Basis of Cancer Phenotypes 15

Page 16: Molecular Biology of Cancer - UC · PDF fileDepartments of Radiation Oncology/Cancer Biology Section Molecular and Cellular Biology ... 2 Molecular Biology of Cancer. are genetic mutations

through the cell cycle. p21waf1, the first discov-ered and best characterized member of theCip/Kip family, is stimulated by the p53 tumorsuppressor protein in response to DNA dam-age and halts cell cycle progression to allow forDNA repair (45). The INK4 family of CKIscontains four member proteins, p16/INK4a,p15/INK4b, p18/INK4c, and p19/INK4d. Un-like the Cip/Kip family, the INK4 proteinshave restricted binding and associate exclu-sively with cdk4/6. Consequently, their princi-pal function is to regulate cyclin D1/cdk4/6 ac-tivity, and therefore, the phosphorylationstatus of the Rb tumor suppressor. p16/INK4ais itself a tumor suppressor that is frequentlymutated in melanoma (46). Indeed, at leastone component of the p16/cyclin D1/Rb path-ways is either mutated or deregulated in somefashion in over 90% of lung cancers, emphasiz-ing the importance of this pathway in regulat-ing tumor cell proliferation.

Transit through the cell cycle is regulatedby two types of controls. In the first type, thecumulative exposure to specific signals, suchas growth factors, is assessed and if the sum ofthese signals satisfies the conditions requiredby the R point, proliferation ensues. In thesecond, feedback controls or checkpoints mon-itor whether the genome is intact and whetherprevious cell cycle steps have been completed.At least five cell cycle checkpoints have beenidentified, two that monitor integrity of theDNA and halt cell cycle progression in eitherG1 or G2, one that ensures DNA synthesis hasbeen completed before mitosis begins, onethat monitors completion of mitosis before al-lowing another round of DNA synthesis, andone that monitors chromosome alignment onthe equatorial plate before initiation of ana-phase. Of these, the two checkpoints thatmonitor integrity of DNA have been the mostextensively studied, and as might be expected,these checkpoints and the genes that enforcethem are critically important for the responsethat cells mount to genotoxic stresses. Abroga-tion of checkpoints leads to genomic instabil-ity and an increased mutation frequency (47).

Progress in elucidating the mechanisms ofcheckpoint function reveals that a number ofcheckpoint genes are frequently mutated inhuman cancers. For example, the p53 tumorsuppressor functions as a cell cycle checkpoint

that halts cell cycle progression in G1 by in-ducing the expression of the p21waf1 gene inthe presence of damaged DNA (45). The p53gene is frequently mutated in human cancersand consequently, most tumor cells lack theDNA damage-induced p53-dependent G1checkpoint, increasing the likelihood that mu-tations will be propagated in these cells. Be-cause p53 also promotes apoptosis, the lack ofp53 in these cells also makes them more resis-tant to the DNA damage-induced apoptosis.Because most chemotherapeutic agents killcells through DNA damage-induced apoptosis,tumor cells with mutant p53 are also moreresistant to conventional therapies (48).

3.5 Apoptosis and Reduced Sensitivityto Apoptosis

Apoptosis is a genetically controlled form ofcell death that is essential for tissue remodel-ing during embryogenesis and for mainte-nance of the homeostatic balance of cell num-bers later in adult life. The importance ofapoptosis to human disease comes from therealization that disruption of the apoptoticprocess is thought to play a role in diverse hu-man diseases ranging from malignancy to neu-rodegenerative disorders. Because apoptosis isa genetically controlled process, much efforthas been spent on identifying these geneticcomponents to better understand the apopto-tic process as well as to identify potential ther-apeutic targets that might be manipulated indisease conditions where disruption of apopto-sis occurs.

Although multiple forms of cell death havebeen described, apoptosis is characterized bymorphological changes including cell shrink-age, membrane blebbing, chromatin conden-sation and nuclear fragmentation, loss ofmicrovilli, and extensive degradation of chro-mosomal DNA. In general, the apoptotic pro-gram can be subdivided into three phases: theinitiation phase, the decision/effector phase,and the degradation/execution phase (Fig.1.7). In the initiation phase, signal transduc-tion pathways that are responsive to externalstimuli, such as death receptor ligands, or tointernal conditions, such as that produced byDNA damage, are activated. During the ensu-ing decision/effector phase, changes in the mi-tochondrial membrane occur that result in

Molecular Biology of Cancer16

Page 17: Molecular Biology of Cancer - UC · PDF fileDepartments of Radiation Oncology/Cancer Biology Section Molecular and Cellular Biology ... 2 Molecular Biology of Cancer. are genetic mutations

disruption of the mitochondrial membrane po-tential and ultimately loss of mitochondrialmembrane integrity. A key event in the deci-sion/effector phase is the release of cyto-chrome c into the cytoplasm and activation ofproteases and nucleases that signal the onsetof the final degradation/execution phase. Animportant concept in understanding apoptosisis that the mitochrondrion is a key target of apo-ptotic stimuli and disruption of mitochondrialfunction is central to subsequent events thatlead to degradation of vital cellular components.

Of the signal transduction pathways thatinitiate apoptosis, the best understood at themolecular level involves the death receptorsincluding Fas/cluster of differentiation 95(CD95), tumor necrosis factor receptor 1(TNFR1), and death receptors 3, 4, and 5 (DR3,4,5) (Fig. 1.8). All death receptors share anamino acid sequence known as the death do-main (DD) that functions as a binding site fora specific set of death signaling proteins. Stim-ulation of these transmembrane receptors canbe induced by interaction with its cognate li-gand or by binding to an agonistic antibody,which results in receptor trimerization and re-cruitment of intracellular death moleculesand stimulation of downstream signalingevents. Here death receptors are classified aseither CD95-like (Fas/CD95, DR4, and DR5)or TNFR1-like (TNF-R1, DR3, and DR6)based on the downstream signaling eventsthat are induced as a consequence of receptoractivation.

Activation of Fas/CD95 leads to clusteringand recruitment of Fas-associated death do-

main (FADD; sometimes called Mort1) to theFas/CD95 intracellular DD (49). FADD con-tains a C-terminal DD that enables it to inter-act with trimerized Fas receptor as well as anN-terminal death effector domain (DED),which can associate with the prodomain of theserine protease, caspase-8. This complex is re-ferred to as the death-inducing signaling com-plex (DISC). As more procaspase-8 is recruitedto this complex, caspase-8 undergoes trans-catalytic cleavage to generate active protease.Activation of TNFR1-like death receptors re-sults in similar events except that the first pro-tein to be recruited to the activated receptor isthe TNFR-associated death domain (TRADD)adaptor protein that subsequently recruitsFADD and procaspase-8. Signaling throughthe TNFR1-like receptors is more complexand includes recruitment of other factors thatdo not interact with Fas/CD95. For example,TRADD also couples with the receptor inter-acting protein (RIP), which links stimulationof TNFR1 to signal transduction mechanisms,leading to activation of nuclear factor-kappa B(NF-�B). Because RIP does not interact withFas/CD95, this class of receptors does not ac-tivate NF-kappa B.

The critical downstream effectors of deathreceptor activation are the caspases, and theseare considered the engine of apoptotic celldeath (50). Caspases are a family of cysteineproteases with at least 14 members. They aresynthesized in the cells as inactive enzymesthat must be processed by proteolytic cleavageat aspartic acid residues. These cleavage sitesare between the N-terminal prodomain, the

Premitochondrialphase

Mitochondrialphase

Postmitochondrialphase

Signaltransduction

Membranepermeabilization

Mitochondrialproteinsreleased

Decision/effectorphase

Initiationphase

Degradation/executionphase

Figure 1.7. Mitochondria-mediated apoptosis. Mitochondria-mediated apoptosis is divided intothree phases. Mitochondrial stress stimulates signal transduction and constitutes the initiationphase. During the second phase, changes in the structure of the mitochondrial membrane make itpermeable to large proteins, allowing the release of cytochrome c and induction of the third and finalphase, during which degradation of cellular proteins occurs.

3 Molecular Basis of Cancer Phenotypes 17

Page 18: Molecular Biology of Cancer - UC · PDF fileDepartments of Radiation Oncology/Cancer Biology Section Molecular and Cellular Biology ... 2 Molecular Biology of Cancer. are genetic mutations

large P20, and small P10 domains. The acti-vated proteases cleave other proteins by recog-nizing an aspartic acid residue at the cleavagesite and are consistent with an auto- or trans-cleavage processing mechanism for activationwhen recruited to activated death receptors.

Importantly, biochemical studies supportthe notion of a caspase hierarchy that consistsof initators and effectors that are activated ina cascade fashion. Initiator caspases such ascaspase-8 and -9 are activated directly by apo-ptotic stimuli and function, in part, by activat-ing effector caspases such as caspase-3, -6, and-7 by proteolytic cleavage. It is the effectorcaspases that result in highly specific cleavageof various cellular proteins and the biochemi-cal and morphological degradation associatedwith apoptosis.

In contrast to death receptor-mediated ap-optosis that functions through a well-defined

pathway, mediators of stress-induced apopto-sis such as growth factors, cytokines, and DNAdamage activate diverse signaling pathwaysthat converge on the mitochondrial mem-brane (51). Many proapoptotic agents havebeen shown to disrupt the mitochondrialmembrane potential (��m), leading to an in-crease in membrane permeability and releaseof cytochrome c into the cytosol. Cytochrome crelease is a common occurrence in apoptosisand is thought to be mediated by opening ofthe permeability transmembrane pore com-plex (PTPC), a large multiprotein complexthat consists of at least 50 different proteins.The cytosolic cytochrome c interacts with ap-optosis activating factor-1 (Apaf-1), dATP/ATP, and procaspase-9 to form a complexknown as the apoptosome. Cytochrome c anddATP/ATP stimulate Apaf-1 self-oligomeriza-tion and trans-catalytic activation of pro-

Ligand(Fas-L)

Disccomplex FADD

Procaspase-8

Procaspase-9

Mitochondrion

Cytochrome c

Mitochondrialstress

BaxBcl-2

Caspase-9

dATP

Apaf-1

Procaspases-3 & 7

Apoptosomecomplex

Procaspases-3 & 7

Activecaspases-3 & 7

Apoptosis

Caspase-8

Receptor(Fas)

Figure 1.8. Apoptosis—receptor-mediated and mitochondrial apoptosis cascades. Trimerization ofthe Fas receptor initiates recruitment of the death domain-containing adaptor protein FADD, whichbinds to procaspase-8 promoting trans-catalytic cleavage of prodomain. Caspase-8 initiates thecaspase cascade by acting on downstream effector caspases 3 and 7. In mitochondria-mediatedapoptosis cytochrome c, release is a key event in apoptosis and is stimulated by Bax and suppressedby Bcl-2. The released cytochrome c binds with Apaf-1 and in conjunction with dATP induces aconformational change in Apaf-1 that permits oligomerization into a �700-kDa complex, which iscalled the apoptosome complex and is capable of recruiting caspases-9, -3, and -7.

Molecular Biology of Cancer18

Page 19: Molecular Biology of Cancer - UC · PDF fileDepartments of Radiation Oncology/Cancer Biology Section Molecular and Cellular Biology ... 2 Molecular Biology of Cancer. are genetic mutations

caspase-9 to the active enzyme. Activecaspase-9 activates effector caspases-3 and -7and leads to the cellular protein degradationcharacteristic of apoptosis.

As release of cytochrome c can have direconsequences for viability of the cell, its re-lease is tightly regulated. Indeed, a whole fam-ily of proteins, of which B-cell lymphoma-2(Bcl-2) is the founding member, that share ho-mology in regions called the Bcl-2 homologydomains are dedicated to regulation of cyto-chrome c release from the mitochondria (52).Both positive regulators (Bax, Bak, Bik, andBid) that promote apoptosis and negative reg-ulators (Bcl-2 and Bcl-XL), which suppress ap-optosis, act by regulating permeability of themitochondrial membrane to cytochrome c.Bcl-2 family members have been found in boththe cytosol and associated with membranes.Bax is normally found in the cytosol, but sub-cellular localization changes during apoptosis.Bax has been shown to insert into the mito-chondrial membrane where, because of itsstructure that is similar to other pore-formingproteins, it is thought to promote release ofcytochrome c. Bcl-2 functions by inhibiting in-sertion of Bax into the mitochondrial mem-brane. Hence, a key factor that determineswhether a cell will undergo apoptosis is theratio of proapoptotic to antiapoptotic Bcl-2family proteins.

Because apoptosis serves to eliminate cellswith a high neoplastic potential, cancer cellshave evolved to evade apoptosis primarilythrough two mechanisms. In the first of these,Bcl-2, which suppresses apoptosis, is overex-pressed. The Bcl-2 oncogene was first identi-fied as a break point in chromosomal translo-cations that frequently occurred in B-cell–derived human tumors. Characterization ofthe rearrangements revealed that the Bcl-2gene is overexpressed by virtue of being placedadjacent to the powerful IgH promoter. Clon-ing of the Bcl-2 gene and overexpression incells of B-cell lineage reduced the sensitivity ofthese cells to apoptosis and allowed them tosurvive under conditions that ordinarilycaused normal cells to die.

The second mechanism that provides can-cer cells with resistance to apoptosis is thesuppression of the Fas receptor. As with otherreceptors, mutations can occur in either the

ligand binding domain or in the intracellulardomain interfering with activation of thedeath signaling pathway. More recently anovel mechanism for suppressing Fas-recep-tor activation has been identified in whichcancer cells synthesize decoy receptors towhich ligands can bind but are unable to in-duce apoptosis (53).

3.6 Increased Genetic Instability

A hallmark of tumor cells is genetic instabilitythat is manifested at the chromosomal level aseither aneuploidy (the gain or loss of one ormore specific chromosomes) or polyloidy (theaccumulation of an entire extra set of chromo-somes). Acquisition of extra chromosomes isone mechanism by which extra copies of agrowth promoting gene can be acquired bycancer cells, providing them with a selectivegrowth advantage. Structural abnormalitiesare also common in advanced tumors that leadto various types of chromosomal rearrange-ments. Translocations and random insertionof genetic material into one chromosome fromanother can place genes that are not normallylocated adjacent to one another in close prox-imity usually leading to abnormal gene ex-pression. Some of these rearrangements areroutinely observed in some cancers such as inBurkitt’s lymphoma where rearrangementsinvolving chromosome 8 and 14 lead to abnor-mal expression of the c-myc protooncogene asa consequence of being placed adjacent to theimmunoglobulin heavy chain promoter.

In chronic myelogenous leukemia (CML),an abnormal chromosome known as the Phil-adelphia chromosome results from a translo-cation involving chromosomes 9 and 22. Thegenes for two unrelated proteins, c-Abl andBcr, a tyrosine kinase, and a GTPase activat-ing protein (GAP), are spliced together, form-ing a chimeric protein that results in a power-ful and constitutively active kinase that drivesproliferation of the cells in which it is ex-pressed.

Other forms of genetic instability includegene amplification. Under normal conditions,all DNA within the cell is replicated uniformlyand only once per cell cycle. However, in can-cer cells some regions of a chromosome canundergo multiple rounds of replication suchthat multiple copies of a growth-promoting

3 Molecular Basis of Cancer Phenotypes 19

Page 20: Molecular Biology of Cancer - UC · PDF fileDepartments of Radiation Oncology/Cancer Biology Section Molecular and Cellular Biology ... 2 Molecular Biology of Cancer. are genetic mutations

gene(s) is obtained. These can result in chro-mosomes with regions of DNA that stain uni-formly during karyotype analysis of a tumorcell or in the production of extrachromosomalDNA-containing bodies known as doubleminute chromosomes. A typical example ofthis type of amplification targets the N-mycgene, which is amplified in �30% of advancedneuroblastomas (54).

More subtle changes at the sequence levelaffecting growth-controlling genes is also com-mon in human tumors. Mutations can occur asa consequence of either defects in DNA repairor decreased fidelity during DNA replication.The components of these pathways are criticalfor maintenance of genome integrity and in-herited mutations in the genes of DNA repairproteins and proteins that repair misrepli-cated DNA explains some inherited cancer-prone syndromes (55).

3.7 Angiogenesis

Without the production of new blood vessels,tumor growth is limited to a volume of a fewcubic millimeters by the distance that oxygenand other nutrients can diffuse through tis-sues. As tumor size increases, intratumoral O2

levels fall and the center of the mass becomeshypoxic, leading to up-regulation of the hyp-oxia inducible factor (HIF1). HIF1 is a het-erodimeric transcription factor composed of aconstitutively expressed HIF-1 beta subunitand an O2 regulatable HIF-1 alpha subunit(56). Under normoxic conditions, levels ofHIF1 are kept low through the actions of theVHL tumor suppressor protein, which func-tions as a ubiquitin ligase that promotes deg-radation through a proteosome mediatedpathway (57). An important transcriptionaltarget of HIF1 is the VEGF growth factor,which in conjunction with other cytokines, in-duces neovascularization of tumors and allowsthem to grow beyond the size limitation im-posed by oxygen diffusion. This increased pro-duction of proangiogenic factors and reduc-tion of anti-angiogenic factors is known as the“angiogenic switch” and is a significant mile-stone in tumorigenesis that leads to the devel-opment of more lethal tumors.

Angiogenesis is the sprouting of capillariesfrom preexisting vessels during embryonic de-velopment and is almost absent in adult tis-

sues with the exception of transient angiogen-esis during the female reproductive cycle andwound healing, and the soluble factor thatplays a critical role in promoting angiogenesisis vascular endothelial growth factor (VEGF)(58). VEGF was first implicated in angiogene-sis when it was identified as a factor secretedby tumor cells, which caused normal bloodvessels to become hyperpermeable (59). Thefollowing evidence supports a role for VEGF intumor angiogenesis.

1. VEGF is present in almost every type ofhuman tumor. It is especially high in con-centration around tumor blood vessels andin hypoxic regions of the tumor.

2. VEGF receptors are found in blood vesselswithin or near tumors.

3. Monoclonal neutralizing antibodies forVEGF can suppress the growth of VEGF-expressing solid tumors in mice. These lackany effect in cell culture where angiogene-sis is not needed.

Ferrara and Henzel (60) identified VEGFas a growth factor capable of inducing prolif-eration of endothelial cells but not fibroblastsor epithelial cells. Inhibition of one of the iden-tified VEGF receptors, FLK1, inhibits thegrowth of a variety of solid tumors (61). Simi-larly, the injection of an antibody to VEGFstrongly suppresses the growth of solid tu-mors of the subcutaneously implanted humanfibrosarcoma cell line HT-1080 (62).

There are several forms of VEGF that seemto have different functions in angiogenesis.These isoforms are VEGF, VEGF-B, VEFG-C,and VEGF-D. VEGF-B is found in a variety ofnormal organs, particularly the heart andskeletal muscle. It can form heterodimerswith VEGF and can affect the availability ofVEGF for receptor binding (63). VEGF-Dseems to be regulated by c-fos and is stronglyexpressed in the fetal lung (64). However, inthe adult it is mainly expressed in skeletalmuscle, heart, lung, and intestine. VEGF-D isalso able to stimulate endothelial cell prolifer-ation (65).

VEGF-C is about 30% homologous toVEGF. Unlike both VEGF and VEGF-B,VEGF-C does not bind to heparin. It is able to

Molecular Biology of Cancer20

Page 21: Molecular Biology of Cancer - UC · PDF fileDepartments of Radiation Oncology/Cancer Biology Section Molecular and Cellular Biology ... 2 Molecular Biology of Cancer. are genetic mutations

increase vascular permeability and stimulatethe migration and proliferation of endothelialcells, although at a significantly higher con-centration than VEGF. VEGF-C is expressedduring embryonal development where lym-phatics sprout from venous vessels (66). It isalso present in adult tissues and may play arole in lymphatic endothelial differentiation.Flt-4, the receptor for VEGF-C, is expressed inangioblasts, veins, and lymphatics during em-bryogenesis, but it is mostly restricted to thelymphatic endothelium in adult tissues. Be-cause of these expression patterns, VEGF-Cand Flt-4 may be involved in lymphangiogen-esis. This is the process of lymphatic genera-tion. Lymphatic vasculature is very importantbecause of its involvement in lymphatic drain-age, immune function, inflammation, and tu-mor metastasis.

Other cytokines and growth factors alsoplay an important role in promoting angiogen-esis. Some of these act directly on endothelialcells, whereas others stimulate adjacent in-flammatory cells. Some can cause migrationbut not division of endothelial cells such asangiotropin, macrophage-derived factor, andTNF�, or stimulate proliferation such as EGF,acidic and basic fibroblast growth factors(aFGF, bFGF), transforming growth factor �(TGF�), and VEGF (67). Tumors secrete thesefactors, which stimulate endothelial migra-tion, proliferation, proteolytic activity, andcapillary morphogenesis (68).

Several angiogenic factors have been iden-tified that can be secreted from tumors. Manyof these are growth factors that are describedas heparin-binding growth factors. Specifi-cally, these include VEGF, FGFs, TGF-�, andthe hepatocyte growth factor (HGF). Thebinding of these factors to heparin sulphateproteoglycans (HSPG) may be a mechanismfor bringing the growth factors to the cell sur-face and presenting them to their appropriatereceptors in the proper conformation. This fa-cilitates the interaction between the growthfactors and receptors. Studies have shownthat tumor growth is adversely affected byagents that block angiogenesis (69) but isstimulated by factors that enhance angiogen-esis (70).

Angiogenesis may be useful as a prognosticindicator. Tumor sections can be stained im-

munohistochemically for angiogenic determi-nants, such as VEGF, to determine the densityof vasculature within the tumor, and there is astrong correlation between high vessel densityand poor prognosis (71). This correlation im-plies a relationship between angiogenesis andmetastasis.

4 CANCER-RELATED GENES

4.1 Oncogenes

Oncogenes are derived from normal hostgenes, also called protooncogenes, that be-come dysregulated as a consequence of muta-tion. Oncogenes contribute to the transforma-tion process by driving cell proliferation orreducing sensitivity to cell death. Historically,oncogenes were identified in four major ways:chromosomal translocation, gene amplifica-tion, RNA tumor viruses, and gene transferexperiments. Gene transfer experiments con-sist of transfecting DNA isolated from tumorcells into normal rodent cells (usually NIH-3T3 cells) and observing any morphologicalchanges. These morphological changes be-came the hallmarks for cell transformation,the process of becoming tumorigenic. As pre-viously discussed, the characteristics of trans-formed cells are as follows: (1) the ability toform foci instead of a monolayer in tissue cul-ture; (2) the ability to grow without adherenceto a matrix, or “anchorage-independentgrowth”; and (3) the ability to form tumorswhen injected into immunologically compro-mised animals.

There are seven classes of oncogenes, clas-sified by their location in the cell and theirbiochemical activity (Table 1.3). All of theseoncogenes have different properties that canlead to cancer. The classes of oncogenes aregrowth factors, growth factor receptors, mem-brane-associated guanine nucleotide-bindingproteins, serine-threonine protein kinases, cy-toplasmic tyrosine kinases, nuclear proteins,and cytoplasmic proteins that affect cell sur-vival.

4.1.1 Growth Factors and Growth FactorReceptors. Cell growth and proliferation aresubject to regulation by external signals thatare typically transmitted to the cell in the

4 Cancer-Related Genes 21

Page 22: Molecular Biology of Cancer - UC · PDF fileDepartments of Radiation Oncology/Cancer Biology Section Molecular and Cellular Biology ... 2 Molecular Biology of Cancer. are genetic mutations

form of growth factors that bind to and acti-vate specific growth factor receptors. Predict-ably, one class of oncogenes consists of growthfactors that can stimulate tumor cell growth.In normal cells and tissues, growth factors areproduced by one cell type that then act on an-other cell type. This is termed paracrine stim-ulation. However, many cancer cells secretetheir own growth factors as well as express thecognate receptors that are stimulated by thosefactors. Because of this autocrine stimulation,cancer cells are less dependent on externalsources of growth factors for proliferation andtheir growth is unregulated. Examples of on-cogenic growth factors include v-sis, which isthe viral homolog of the platelet-derivedgrowth factor (PDGF) gene. PDGF stimulates

the proliferation of cells derived from connec-tive tissue such as fibroblasts, smooth musclecells, and glial cells. Thus, tumors caused byexcess stimulation by v-sis include fibrosarco-mas and gliomas.

The receptors that interact with growthfactors are also another large family of onco-genes. Growth factor receptors are composedof three domains: an extracellular domainthat contains the ligand binding domain thatinteracts with the appropriate growth factor, ahydrophobic transmembrane domain, and acytoplasmic domain that typically contains akinase domain that can phosphorylate ty-rosine residues in other proteins. Hence, thesereceptors are frequently referred to as recep-tor tyrosine kinases (RTK). It is this kinase

Table 1.3 Oncogenes

Oncogenes Protein Function Neoplasm(s)

Growth Factorssis Platelet-derived growth factor fibrosarcomaint-2 Fibroblast growth factor breasttrk Nerve growth factor neuroblastoma

Growth Factor Receptorserb-B1 Epidermal growth factor receptor squamous cell

carcinomaerb-B2/HER2/neu Heregulin breast carcinomafms Hematopoietic colony stimulating factor sarcomaros Insulin receptor astrocytoma

Tyrosine kinasesbcr-abl Tyrosine kinase chronic myelogenous

leukemiasrc Tyrosine kinase colonlck Tyrosine kinase colon

Serine-Threonine protein kinasesraf Serine-threonine kinase sarcomamos Serine-threonine kinase sarcoma

Guanine nucleotide binding proteinsH-ras GTPase melanoma; lung,

pancreasK-ras GTPase leukemias; colon,

lung, pancreasN-ras GTPase carcinoma of the

genitourinarytract and thyroid;melanoma

Cytoplasmic proteinsbcl-2 Anti-apoptotic protein non-Hodgkin’s B-cell

lymphomaNuclear proteins

myc Transcription factor Burkitt’s lymphomajun Transcription factor (AP-1) osteosarcomafos Transcription factor (AP-1) sarcoma

Molecular Biology of Cancer22

Page 23: Molecular Biology of Cancer - UC · PDF fileDepartments of Radiation Oncology/Cancer Biology Section Molecular and Cellular Biology ... 2 Molecular Biology of Cancer. are genetic mutations

activity that is essential to the intracellularsignaling that is stimulated by an activatedreceptor and in all oncogenic receptors muta-tions that lead to constitutive intracellularsignaling promote unregulated cellular prolif-eration. RTKs can become oncogenically acti-vated by mutations in each of the protein do-mains. Genetic mutations that result in theproduction of an epidermal growth factor re-ceptor (EGFR) lacking the extracellular li-gand binding domain leads to constitutive sig-naling. This oncogenic EGFR is known aserb-B1 (Fig. 1.9).

Normally, EGF binds to the extracellularportion of the EGFR and causes dimeriza-tion of the intracellular part of the receptorand association with adaptor proteins, Sonof Sevenless (SOS), and growth factor recep-tor binding protein 2 (Grb 2). These proteinsinteract through src-homology (SH) do-

mains SH2 and SH3, respectively. Throughan unknown mechanism, the SOS-Grb 2complex activates the oncogene ras. Ras in-duces an intracellular cascade of kinases topromote proliferation. These signaling cas-cades become constitutive when the extra-cellular portion of the EGFR becomes trun-cated, as in the case of erb-B1. Oncogenicactivation of a related RTK, erb-B2, occursas a consequence of a single point mutationthat falls within the transmembrane regionof this receptor (72). This mutated receptoris frequently found in breast cancers. Fi-nally, mutations in the cytoplasmic kinasedomain can also cause constitutive activityleading to constitutive signaling.

4.1.2 G Proteins. In many cases, signalingthat is initiated by growth factors activatingtheir receptors passes next to membrane asso-

Akt/PKB

JNKERK

GF

P

P

Grb2

GDP

SOS

Ras Raf

MEK

PKC

Growthfactorreceptor

SEK

GTP

NucleusCell survival

Proliferation

PI-3K

Bad

Figure 1.9. Ras signaling pathway. Growth factor (GF) binds to its receptor and initiates dimeriza-tion and autophosphorylation. Grb2 interacts with SOS, which activates ras by promoting the GTP-bound form. Ras recruits Raf to the plasma membrane and initiates the Raf/MAPK signaling cascade.Protein kinase C also stimulates this pathway as well as another cascade of stress-activated kinases(SEK/JNK). Both of these signaling pathways promote cell proliferation by stimulating the transcrip-tion of genes like cyclooxygenase-2, activator protein-1, and nuclear factor-�B. Ras also signalsphosphoinositol-3-kinase and Akt/protein kinase B for cell survival.

4 Cancer-Related Genes 23

Page 24: Molecular Biology of Cancer - UC · PDF fileDepartments of Radiation Oncology/Cancer Biology Section Molecular and Cellular Biology ... 2 Molecular Biology of Cancer. are genetic mutations

ciated guanine nucleotide-binding proteins,which when activated by mutation, constituteanother class of oncogenes. The prototypicalmember of this family of oncogenes is the rasoncogene. There are three ras genes in thisfamily of oncogenes, which include H-ras, K-ras, and N-ras. These genes differ in their ex-pression patterns in different tissues. All havebeen found to have point mutations in humancancers including liver, colon, skin, pancre-atic, and lung cancers, which lead to constitu-tive signaling of genes involved in prolifera-tion, cell survival, and remodeling of the actincytoskeleton. Ras is a small molecular weightprotein that is post-translationally modifiedby attachment of a farnasyl fatty acid moietyto the C-terminus. Because this post-transla-tional modification is essential for activity ofthe ras oncogenes, this process has become atarget for drug development aimed at interfer-ing with ras activity (73).

Ras binds both guanosine 5�-triphosphate(GTP) and guanosine 5�-diphosphate (GDP)reversibly but is only in the activated state andcapable of signaling when bound to GTP. Theactivated, GTP-bound form of ras signals a va-riety of mitogen-induced and stress-inducedpathways, leading to transcription of genesnecessary for cell growth and proliferation(74). Mitogens such as growth factors can ac-tivate ras through the epidermal growth fac-tor receptor, and stress factors affecting rasinclude ultraviolet light, heat, and genotoxins.Guanine nucleotide exchange factors (GEFs)foster ras activation by promoting the ex-change of GDP for GTP. In contrast, GTPaseactivating proteins (GAPs) suppress ras activ-ity by promoting GTP hydrolysis by ras, re-sulting in the GDP-bound inactive form of ras(75). Importantly, because GAPs function tosuppress cell proliferation, they can bethought of as tumor suppressors. Indeed, theneurofibromatosis gene, NF-1, is a GAP thatacts as a tumor suppressor gene and can beinherited in a mutated and nonfunctionalform giving rise to the Von Recklinghausenneurofibromatosis or neurofibromatosis type1 cancer syndrome (76).

4.1.3 Serine/Threonine Kinases. Once acti-vated, ras then transmits the growth signal toa third class of signaling molecules that is

comprised of the serine/thereonine kinases.The best studied of these serine-threonineprotein kinases is the raf oncogene, which isactivated when it is recruited to the plasmamembrane by ras (77). Raf then initiates a cas-cade of mitogen-induced protein kinases(MAPKs), which culminate in the nucleuswith the activation of genes containing Elk-1transcription factor binding sites. Raf can alsodirectly activate protein kinase C, which sig-nals another set of kinases that phosphorylatethe c-jun transcription factor.

Another ras effector gene is phosphoinosi-tol 3-kinase (PI-3K), which initiates a signal-ing pathway for cell survival (78). PI-3Kphosphorylates phosphatidalinositol (3,4,5)-triphosphate (PtdIns-3,4,5-P3), an importantintracellular second messenger, thus aiding inthe transmission of signals for proliferation tothe nucleus. PI-3K consists of a catalytic sub-unit, p110, and a regulatory subunit, p85, andthere are five isoforms of each subunit. PI-3Kphosphorylates protein kinase B (Akt/PKB)on serine and threonine residues, which inturn modulate cellular processes like glycoly-sis and translation initiation and elongation.Akt/PKB also phosphorylates Bad, a pro-apop-totic protein. When Bad is phosphorylated, itis sequestered by the 14-3-3 protein, renderingit incapable of binding to the anti-apoptoticprotein, bcl-2, and thus, results in apoptosis.Akt’s phosphorylation of Bad serves to inhibitapoptosis and promote cell survival. This hasdeleterious effects for the organism becausetumor cells are not permitted to undergo apo-ptosis and will survive and divide.

PI-3K has been linked to the developmentof colon cancer by a study showing that geneticinactivation of the p110gamma catalytic sub-unit of PI-3K leads to the development of in-vasive colorectal adenocarcinomas in mice(79). This pathway is not completely separatefrom the Raf/MAPK pathway, because Akt hasbeen found to inhibit Raf activity. In fact, noneof the aforementioned ras-mediated pathwaysoperate completely independently; there aremultiple examples of crosstalk between thesesignaling pathways.

4.1.4 Nonreceptor Tyrosine Kinases. In ad-dition to growth factor receptors, other nonre-ceptor kinases target protein tyrosines for

Molecular Biology of Cancer24

Page 25: Molecular Biology of Cancer - UC · PDF fileDepartments of Radiation Oncology/Cancer Biology Section Molecular and Cellular Biology ... 2 Molecular Biology of Cancer. are genetic mutations

phosphorylation and can become activated asoncogenes. Indeed, one of the first oncogenesto be discovered, src, is the best characterizedmember of a family of proteins that have on-cogenic potential. The src family of proteinsare post-translationally modified by attach-ment of a myristate moiety to the N-terminus,which enables association with the plasmamembrane. The members of the src family ofproteins exhibit 75% homology at the aminoacid level with the greatest degree of similarityfound in three regions that have been labeledsrc homology domains 1, 2, and 3 (e.g., SH1,SH2, and SH3). The SH1 domain encompasesthe domain that contains kinase activity. TheSH2 and SH3 domains are located adjacent toand N-terminal to the kinase domain andfunction to promote protein/protein interac-tions. The SH2 domain binds with phosphor-ylated tyorsines, whereas the SH3 domain hasaffinity for the proline rich regions of proteins.Importantly, SH2 and SH3 domains are foundin a large number of other proteins that areinvolved in intracellular signaling and thathave oncogenic potential, and the structure ofthese domains are strongly conserved. Be-cause SH2 and SH3 domains serve to potenti-ate signal transduction, they have also becometargets for drug discovery programs aimed atdisrupting the constitutive signaling gener-ated by oncogenic activity (80).

A second oncogenic protein tyrosine kinaseof considerable clinical importance is the Bcr-Abl oncogene. The Bcr-Abl protein is a chi-meric fusion protein formed by a reciprocaltranslocation involving chromosomes 9 and22. This chromosomal rearrangement is diag-nostic for the hematopoietic malignancy,chronic myelogenous leukemia (CML), andthe rearranged chromosome is known as thePhiladelphia chromosome (81). The c-Ablgene maps to chromosome 9 and is a tyrosinekinase, whereas the BCR gene is now knownto be GTPase-activating protein (GAP), whichwhen fused to Abl results in an unregulatedtyrosine kinase that functions to promote cel-lular proliferation (82). The bcr-abl protein in-teracts with SH2 domains on Grb 2 and relo-cates to the cytoskeleton and initiates rassignaling, a primary mode of tumorigenic po-tential. Bcr-abl reduces growth factor depen-dence, alters adhesion properties, and en-

hances viability of CML cells. Consequently,the kinase activity of Bcr-Abl is a primary fac-tor in stimulating the proliferation of CMLcells, and therefore, has become the target fordrug therapies aimed at combating this can-cer. Indeed, the drug STI571 has been spectac-ularly successful in the clinic at causing remis-sion of this disease (83).

4.1.5 Transcription Factors as Oncogenes.Another class of oncogenes are those that en-code nuclear proteins, or transcription factors.Two examples of this class of oncogenes areAP-1 and c-myc. Activator protein-1 (AP-1)consists of Fos family members (c-fos, fos B,Fra 1, and Fra 2) and Jun family members(c-jun, jun B, and jun D), which can dimerizethrough a lucine rich protein/protein interac-tion domain known as the leucine zipper (84).Fos-jun heterodimers are the most active, jun-jun homodimers are weakly active, and fos-foshomodimers form only in extremely rare cir-cumstances. These dimers bind to AP-1 DNAbinding sites, which are also called the tumorpromoter TPA-responsive element (TRE) orglucocorticoid response element (GRE). AP-1can be activated by ionizing and ultravioletirradiation, DNA damage, cytokines, and oxi-dative and cellular stresses (85).

AP-1 has several functions in the cell, in-cluding the promotion of cell proliferation andmetastasis. AP-1 is a nuclear target for growthfactor-induced signaling such as the afore-mentioned EGFR-mediated kinase cascade.AP-1–regulated genes include genes necessaryfor metastasis, and invasion like the MMPsmatrilysin and stromelysin, as well as collage-nase two proteins that aid in cell migrationthrough connective tissue.

Deregulation of c-myc often occurs eitherby gene rearrangement or amplification in hu-man cancers. Here again the hematologic can-cers are instructive. In Burkitt’s lymphoma, afrequent reciprocal translocation betweenchromosomes 8 and 14 leads to juxtaposition-ing of the myc gene adjacent to the Ig heavychain promoter/enhancer complex, causinguncontrolled expression and production of themyc protein (86). Translocations betweenchromosomes 2 and 8 and between 8 and 22also occur and involve other immunoglobulin

4 Cancer-Related Genes 25

Page 26: Molecular Biology of Cancer - UC · PDF fileDepartments of Radiation Oncology/Cancer Biology Section Molecular and Cellular Biology ... 2 Molecular Biology of Cancer. are genetic mutations

producing gene complexes. In all cases theoverproduction of myc results in uncontrolledcell proliferation.

Myc overexpression also occurs in solid tu-mors, but is usually the result of gene amplifi-cation (87). The oncogenic potential of c-mychas been studied most widely as it pertains tothe development of colon cancer. Both c-mycRNA and protein are overexpressed at theearly and late stages of colorectal tumorigene-sis. The cause for this overexpression is stillunknown, but a strong possibility may be thatit is regulated by the APC pathway. The APCtumor suppressor gene is mutated in approxi-mately 90% of colorectal tumors, both spo-radic and inherited forms. APC will be dis-cussed in detail in the “tumor suppressor”section of this chapter.

He et al. (88) found that when APC expres-sion was induced in stably transfected APC�/�

colon cancer cells (using an inducible metallo-thionine promoter linked to the APC gene),they observed a time-dependent decrease inthe RNA and protein levels of c-myc. This sug-gested that c-myc may be regulated by APCthrough the �-catenin/T-cell factor-4 (Tcf-4)transcription complex. They also showed thatconstitutive expression of mutant �-catenin(mutated so that it is insensitive to APC) inembryonic kidney cells resulted in a signifi-cant increase of c-myc expression. Analysis ofthe c-myc gene revealed two possible Tcf-4transcription factor binding sites. Mobilityshift assays demonstrated that Tcf-4 binds toboth of the potential binding sites, leading toc-myc gene expression. Expression of domi-nant-negative Tcf-4 in HCT116 (mutant�-catenin) or SW480 (mutant APC) reducedendogenous levels of c-myc (88).

The c-myc protein binds to DNA throughits basic, helix-loop-helix/leucine zipper do-main. Many target genes of c-myc have beenidentified that are involved in cell growth andproliferation. Some of these genes includeODC, cell cycle genes cyclins A, E, and D1, aswell as cdc2, cdc25, eukaryotic initiation fac-tor 4E (eIF4E), heat shock protein 70 (hsp70),and dihydrofolate reductase. Overexpressionof c-myc may therefore affect the transcrip-tion of these genes, thus promoting hyperpro-liferation and tumorigenesis.

C-myc is also found to be amplified in pro-myelocytic leukemia and small cell lung can-cer. The c-myc protein requires dimerizationwith Max to initiate transcription, and Maxhomodimers serve as an antagonist of tran-scription. The formation of Mad-Max dimersalso suppresses transcription. It is also inter-esting to note that the full oncogenic potentialof c-myc relies on cooperation with other on-cogenes like ras.

4.1.6 Cytoplasmic Proteins. Bcl-2 is an ex-ample of a cytoplasmic oncogene that has anti-apoptotic potential. Increased production ofbcl-2 protein is seen in a variety of tumor typesand is associated with poor prognosis in carci-nomas of the colon and prostate. The functionof bcl-2 is explained in detail in the “apopto-sis” section of this chapter.

4.2 Tumor Suppressor Genes

In contrast to oncogenes, tumor suppressorgenes can directly or indirectly inhibit cellgrowth. Those that directly inhibit cell growthor promote cell death are known as “gatekeep-ers” and their activity is rate limiting for tu-mor cell proliferation. Hence, both copies ofgatekeeper tumor suppressors must be func-tionally eliminated for tumors to develop. Thischaracteristic requirement is a hallmark of tu-mor suppressor genes. Mutations that inacti-vate one allele of a gatekeeper gene can beinherited through the germline, which in con-junction with somatic mutation of the remain-ing allele, leads to cancer predisposition syn-dromes. For example, mutations of the APCgene lead to colon tumors. Somatic mutationsthat inactivate both gatekeeper alleles occurin sporadic tumors.

Those tumor suppressor genes that do notdirectly suppress proliferation, but function topromote genetic stability are known as “care-takers.” Caretakers function in DNA repairpathways and elimination of caretakers re-sults in increased mutation rates. Because nu-merous mutations are required for the fulldevelopment of a tumor, elimination of care-taker tumor suppressors can greatly acceler-ate tumor progression. As with gatekeepers,mutations can be inherited through the germ-line and can give rise to cancer predispositionsyndromes. An example of a caretaker gene is

Molecular Biology of Cancer26

Page 27: Molecular Biology of Cancer - UC · PDF fileDepartments of Radiation Oncology/Cancer Biology Section Molecular and Cellular Biology ... 2 Molecular Biology of Cancer. are genetic mutations

MSH2, which functions in the mismatch DNArepair system, and inherited mutations in thisgene gives rise to the hereditary nonpolyposiscolorectal cancer (HNPCC) syndrome (Table1.4).

4.2.1 Retinoblastoma. Retinoblastoma(Rb) is a childhood disease. There are both he-reditary and nonhereditary forms of the dis-ease. Approximately 60% of patients developthe nonhereditary form and present with uni-lateral tumor development (one eye is af-fected). About 40% of Rb patients have a germ-line mutation that predisposes them to thedisease. Of these patients, 80% of the cases arebilateral, 15% are unilateral, and about 5% areasymptomatic carriers of the mutation. It is anautosomal dominant trait and is caused bymutations in the Rb gene on chromosome 13.Abnormalities of the Rb gene have also beenseen in breast, lung, and bladder cancers.

Retinoblastoma arises when both of the Rballeles are inactivated. In the inherited form,one parental chromosome carries a defect(most often a deletion) at the Rb locus. A sec-ond somatic mutation must occur in retinalcells to cause the loss of the other (normal) Rballele. In sporadic cases, both of the parentalchromosomes are normal and both Rb allelesare lost as a result of individual somatic muta-tions. Approximately one-half of all retino-blastoma cases show a deletion at the Rb locus.The locus is very large, �150 kb, and there-

fore may be more susceptible to mutations be-cause it is such a large target.

Rb was the first human tumor suppressorgene identified, and the loss of RB proteinfunction leads to malignancy. The RB proteinis localized in the nucleus where it is eitherphosphorylated or unphosphorylated (Fig.1.10). When unphosphorylated, RB binds tothe E2F transcription factor and preventstranscriptional activation of E2F target genes.This normally occurs during the M and earlyG1 phases of the cell cycle. During late G1, S,and G2 phases, RB is phosphorylated. Whenphosphorylated, RB can no longer bind toE2F. This release from inhibition allows E2Fto activate transcription of S-phase genes andthe cell cycle progresses. When loss of RB func-tion occurs because of various mutations inthe Rb gene, the cell cycle becomes deregu-lated, and uncontrolled cell division results.This is because RB can no longer bind to andinhibit E2F. Therefore, the transcription fac-tor can constitutively activate its target genes.This ultimately leads to tumor development(89).

4.2.2 p53. The p53 tumor suppressor is ac-tivated in response to a wide variety of cellularstresses including DNA damage, ribonucle-otide depletion, redox modulation, hypoxia,changes in cell adhesion, and the stresses cre-ated by activated oncogenes. The p53 proteinfunctions as a transcription factor that, when

Table 1.4 Tumor Suppressor Genes

TS Gene Protein Function Neoplasm(s)

APC cell adhesion colonBRCA 1 transcription factor breast and ovaryBRCA 2 DNA repair breast and ovaryCDK4 cyclin D kinase melanomahMLH1 DNA mismatch repair HNPCCa

hMSH2 DNA mismatch repair HNPCChPMS1 DNA mismatch repair HNPCChPMS2 DNA mismatch repair HNPCCMEN1b Ret receptor thyroidNF1 GTPase neuroblastomap53 transcription factor colon, lung, breastRb cell cycle checkpoint retinoblastomaWT-1 transcription factor childhood kidney

aHereditary non-polyposis colon cancer.bMultiple endocrine neoplasia.

4 Cancer-Related Genes 27

Page 28: Molecular Biology of Cancer - UC · PDF fileDepartments of Radiation Oncology/Cancer Biology Section Molecular and Cellular Biology ... 2 Molecular Biology of Cancer. are genetic mutations

activated, stimulates the expression of a vari-ety of effectors that bring about growth arrest,promote DNA repair, and stimulate cell deathby apoptosis. Collectively these activities actto maintain genomic stability. Elimination ofp53 function leads to increased rates of muta-tion and resistance to apoptosis. Thus, p53 sitsat the crux of several biochemical pathwaysthat are disrupted during tumorigenesis. Con-sequently, mutations in p53 are the most fre-quent genetic change encountered in humancancers.

p53 activity can be eliminated by at leastthree mechanisms. The most common eventthat leads to a nonfunctioning protein is mu-tation of the p53 gene, which occurs in about50% of all sporadic human tumors. As withother tumor suppressors, mutations can occurin somatic tissues or can be inherited throughthe germline. Inherited p53 mutations giverise to the Li-Fraumeni syndrome in whichaffected individuals develop bone or soft-tis-sue sarcomas at an early age. In addition, non-mutational inactivation of p53 can occur in thepresence of viral transforming antigens. For

example, the simian virus 40 (SV40) large Tantigen binds with p53 and forms an inactivecomplex, whereas the papilloma virus E6 pro-tein eliminates p53 by causing premature deg-radation of the protein through the 26S pro-teosome. Clearly, the interaction betweenthese transforming antigens and p53 is criticalbecause viral antigens that are incapable ofdoing so lose their transforming ability. Thethird mechanism by which p53 activity can beeliminated is by cytoplasmic sequestration.p53 that is unable to enter the nucleus cannotinduce the expression of downstream effectorgenes that are necessary for mounting the cel-lular response to genotoxic stress.

Activation of p53 by ionizing radiation (IR)and other DNA damaging agents involves acomplex set of interdependent post-transla-tional modifications that control protein/protein associations, protein turnover, andsubcellular localization. Under normal condi-tions, levels of p53 are kept minimal by ubiq-uitination and proteosome-mediated degrada-tion that contributes to the short half-life(3–20 min) of the protein. A key player in

E2FRb

Transcription of E2F targetgenes is inhibited

Rb P E2F

E2F

Transcription of S phase genes

Cell cycle progression

M-phase and early G1 Late G1, S, and G2 phases

Cyclin D/Cdk4

Rb sequesters and inhibits the function of E2F Phosphorylated Rb releases E2Fwhich is then free to bind to its target genes

+

Figure 1.10. Cell cycle control by the retinoblastoma (Rb) tumor suppressor protein. Unphosphor-ylated Rb negatively regulates progression into the S phase of the cell cycle by binding to the E2Ftranscription factor. In this complex, E2F is prevented from activating transcription of its targetgenes. During late G1, Rb is phosphorylated by the cyclin D/Cdk4 complex and can no longer seques-ter the E2F transcription factor. E2F then binds to its target S-phase genes, promoting their tran-scription and allowing the cell cycle to progress.

Molecular Biology of Cancer28

Page 29: Molecular Biology of Cancer - UC · PDF fileDepartments of Radiation Oncology/Cancer Biology Section Molecular and Cellular Biology ... 2 Molecular Biology of Cancer. are genetic mutations

maintenance of low p53 levels is mdm2. Mdm2performs this function by interacting with p53at its N-terminus and targets p53 for proteo-some-mediated degradation. Exposure to IRresults in a series of, as yet incompletely un-derstood, phosphorylation events in p53’s N-terminus, which inhibits Mdm2 binding andresults in increased intracellular p53 levels.Mdm2 and p53 function in a feedback loopwhere activated p53 stimulates the expressionof Mdm2, which in turn reduces the durationof up-regulated p53 activity. Overexpressionof Mdm2 suppresses p53 by preventing its ac-cumulation in response to DNA damage. Con-sequently, Mdm2 can function as an onco-gene that acts in much the same way as thepapilloma virus E6 protein. In fact, Mdm2 isoverexpressed in some tumors such asosteosarcomas.

The p53 protein can be divided into threestructural domains that are essential for tu-mor suppressor function. The N-terminusconsists of a transactivation domain that in-teracts with various basal transcription fac-tors and cellular and viral proteins that mod-ify its function. The central domain containsthe sequence specific DNA binding activity.Most mutations in the p53 gene fall withinthis domain that disrupts the structure of thisregion and eliminates DNA binding activity.The importance of DNA binding is empha-sized by the fact that mutations accumulatepreferentially in several amino acids that areinvolved in directly contacting DNA. The C-terminus has been assigned several activitiesincluding non-specific DNA binding activity,acting as a binding site for other p53 mole-cules, and formation of p53 tetramers, andfunctioning as a pseudosubstrate domain thatoccludes the central DNA binding domain.

Because of the frequency with which p53 ismutated in human tumors, much attentionhas been directed at developing methods thatcompensate for the loss of wild-type functionor can reactivate wild-type p53 activity in mu-tant proteins. For example, strategies aimedat manipulating the conformation of mutantproteins have led to the discovery that pep-tides that bind the C-terminus can reactivatewild-type function in some mutant proteins.Strategies that take advantage of the vastknowledge of virus biology and p53 function

have lead to the construction of viral vectorsthat can introduce a wild-type p53 into tumorcells. One clever approach takes advantage ofthe fact that adenoviruses with a defectiveE1B 55K protein cannot replicate in normalhuman cells. For adenoviruses to replicate incells, they must suppress p53 activity, whichfunctions to limit the uncontrolled DNA repli-cation that is required for production of virusgenomes. However, adenoviruses with a defec-tive E1B 55K gene can replicate in tumor cellsbecause they lack a functional p53. Thus,these viruses kill tumor cells specifically andleave normal cells untouched (90).

4.2.3 Adenomatous Polyposis Coli. The tu-mor suppressor gene, APC, is mutated in al-most 90% of human colon cancers and 30% ofmelanoma skin cancers. The inherited loss ofAPC tumor suppressor function results in fa-milial adenomatous polyposis (FAP). FAP pa-tients develop hundreds to thousands of colonpolyps by their second or third decade of life.By age 40, one or two of these polyps usuallydevelops into a malignant carcinoma, andthus, many of these patients choose to have acolectomy to prevent carcinoma formation.Mutations in APC occur in the majority of spo-radic colon cancers too.

APC mutation is an early event in coloncarcinogenesis, and is therefore, considered tobe the initiating event. Loss of this tumor sup-pressor gene results in constitutive activity ofthe oncogene, c-myc, through an intricate col-lection of protein-protein interactions. Briefly,APC interacts with other cellular proteins, in-cluding the oncogene �-catenin (Fig. 1.11).Axin, an inhibitor of Wnt signaling, forms acomplex with glycogen synthase kinase 3�(GSK3�), �-catenin, and APC and stimulatesthe phosphorylation of �-catenin by GSK3�,thus causing down-regulation of gene expres-sion mediated by �-catenin/Tcf complexes(91). Dissociation of the axin, GSK3�, �-cate-nin, and APC complex by Wnt family membersleads to stabilization of �-catenin and activa-tion of Tcf-mediated transcription. Deletion ofAPC alleles, or mutations causing truncationsin APC that influence its interaction with�-catenin, also leads to stabilization of �-cate-nin and activation of Tcf/lymphoid enhancingfactor (Lef)–dependent gene expression. At

4 Cancer-Related Genes 29

Page 30: Molecular Biology of Cancer - UC · PDF fileDepartments of Radiation Oncology/Cancer Biology Section Molecular and Cellular Biology ... 2 Molecular Biology of Cancer. are genetic mutations

least one member of the Tcf/Lef family of tran-scriptional activators has been identified inhuman colon mucosal tissues. This member istermed hTcf-4. Several target genes for Tcf/Lef have been identified, including the c-myconcogene. Overexpression of wild-type APCcDNA in human colon tumor-derived HT29cells, which lack a normal APC allele, causesdown-regulation of c-myc transcription. Up-regulation of �-catenin in cells expressing nor-mal APC alleles causes increased c-myc ex-pression. Thus, wild-type APC serves tosuppress c-myc expression. Either normal reg-ulation by Wnt signaling, or mutation/dele-tion of APC, activates c-myc expression. Inmany colon cancers, the APC gene is not nec-essarily mutated, but the mutation in thepathway is found in �-catenin, which yieldsthe same constitutive signaling from thepathway.

APC regulates the rates of proliferationand apoptosis by several different mecha-nisms. Wild-type APC is important for cy-toskeletal integrity, cellular adhesion, andWnt signaling. APC plays a role in the G1/Stransition of the cell cycle by modulating ex-pression levels of c-myc and cyclin D1. Wild-type, full length APC is also important inmaintaining intestinal cell migration up thecrypt and inducing apoptosis.

4.2.4 Phosphatase and Tensin Homologue.The phosphatase and tensin homologue(PTEN) or mutated in multiple advanced can-cers (MMAC) tumor suppressor gene was firstidentified in the most aggressive form of braincancer, glioblastoma multiform. PTEN also ismutated in a significant fraction of endome-trial carcinomas, prostate carcinomas, andmelanomas. PTEN’s primary functions as atumor suppressor gene are the induction ofcell cycle arrest and apoptosis (92). PTEN is adual-specificity phosphatase, meaning that itcan dephosphorylate proteins on serine, thre-onine, and tyrosine residues. It specifically de-phosphorylates PtdIns-3,4,5-P3, antagonizingthe function of PI-3K. PTEN, therefore, actsas a negative regulator of Akt activation. Be-cause Akt can suppress apoptosis by the phos-phorylation of the pro-apoptotic protein Bad,PTEN can induce apoptosis of mutated orstressed cells to prevent tumor formation.

In addition to modulating apoptosis, PTENplays a role in angiogenesis. PTEN suppressesthe PI-3K-mediated induction of blood vesselgrowth factors like VEGF. EGF and ras act toinduce genes regulated by the hypoxia-in-duced factor (HIF-1), which is blocked byPTEN activity. PTEN also inhibits cell migra-tion and formation of focal adhesions whenoverexpressed in glioblastoma cell lines, sug-gesting that it helps to inhibit metastasis aswell (93).

PTEN also inhibits signaling from the in-sulin growth factor receptor (IGF-R). Insulinreceptor substrates-1/2 (IRS-1/2) are dockingproteins that are recruited by the insulin re-ceptor and in turn, recruit PI-3K for signaltransduction. The tumor suppressor functionof PTEN helps to prevent aberrant signalingwhen insulin binds to its cell surface receptor.

4.2.5 Transforming Growth Factor-�.Transforming growth factor-� (TGF-�) isgrowth stimulatory in endothelial cells butgrowth inhibitory for epithelial cells, render-ing it a tumor suppressor gene in epithelial-derived cancers. The TGF-� family of growthfactors binds to two unique receptors, TGF-�type I and type II. Tumor cells lose their re-sponse to the growth factor and mutations inthe receptors also contribute to carcinogenesis.Ligand binding to the TGF-� receptors causesintracellular signaling of other tumor suppres-sor genes, the Smad proteins. Smads help to ini-tiate TGF-�-mediated gene transcription.

TGF-�1 normally inhibits growth of hu-man colonic cells, but in the process of becom-ing tumorigenic, these cells obtain a decreasedresponse to the growth inhibitory actions ofTGF-�. TGF-�1 also serves as an inhibitor ofimmune surveillance (94). TGF-�1 indirectlysuppresses the function of the immune systemby inhibiting the production of TNF-� and byinhibiting the expression of class II major his-tocompatibility complex (MHC) molecules.TGF-�1 also promotes tumor progression bymodulating processes necessary for metasta-sis such as degradation of the extracellularmatrix, tumor cell invasion and VEGF-medi-ated angiogenesis.

The TGF-� receptor type II (T�RII) is mu-tated in association with microsatellite insta-bility in most colorectal carcinomas (95). As

Molecular Biology of Cancer30

Page 31: Molecular Biology of Cancer - UC · PDF fileDepartments of Radiation Oncology/Cancer Biology Section Molecular and Cellular Biology ... 2 Molecular Biology of Cancer. are genetic mutations

WNT DSH

MutantAPC

WNT DSH

Target genes

Tcf-4

Nucleus

E-cadherin

26Sproteasome

Axin

APC

E-cadherin

Axin

Normal APC

Mutant APC

Nucleus

Target genes

Tcf-4

Figure 1.11. The APC signalingpathway. In a normal cell, APC forms acomplex with axin, GSK-3�, and�-catenin. This promotes proteosomaldegradation of �-catenin and preventstranscription of �-catenin/Tcf4 targetgenes. When APC is mutated, themulti-protein complex cannot formand �-catenin is not degraded. Instead,�-catenin is translocated to the nu-cleus where it binds with Tcf4 to acti-vate transcription of various targetgenes. Some of the known targetgenes, like c-myc and cyclin D1, playimportant roles in cell proliferation.

4 Cancer-Related Genes 31

Page 32: Molecular Biology of Cancer - UC · PDF fileDepartments of Radiation Oncology/Cancer Biology Section Molecular and Cellular Biology ... 2 Molecular Biology of Cancer. are genetic mutations

many as 25% of colon cancers have missensemutations in the kinase domain of this recep-tor. A missense mutation in the kinase domainof the T�RI has also been identified in meta-static breast cancer. It was also found that theexpression of the TGF-�2 receptor is sup-pressed in metastatic oral squamous cell car-cinomas compared with the primary tumor.

4.2.6 Heritable Cancer Syndromes. Thereare several known inheritable DNA repair-de-ficiency diseases. Four of these are autosomalrecessive diseases and include Xeroderma pig-mentosum (XP), ataxia telangiectasia (AT),Fanconi’s anemia (FA), and Bloom’s syn-drome (BS). XP patients are very sensitive toUV light and have increased predisposition toskin cancer (approximately 1000-fold) (96).AT patients exhibit a high incidence of lym-phomas, and the incidence of lymphoma devel-opment is also increased for both FA and BSpatients.

HNPCC arises due to a defect in mismatchrepair (MMR). The incidence of HNPCC is of-ten quoted as 1–10% of all colorectal cancers(97). It is an autosomal dominant disease andresults in early onset of colorectal adenocarci-noma. Many of these tumors demonstrate mi-crosatellite instability and are termed replica-tion error positive (RER�). Endometrial andovarian cancers are the second and third mostcommon cancers in families with the HNPCCgene defect.

The most common mutations in HNPCCare in the mismatch repair genes, MSH2 andMLH1 (�80%) (98). The mismatch repair sys-tem normally corrects errors of 1–5 base pairsmade during replication. Therefore, defects inthis system result in many errors and createmicrosatellite instability. A suggested modelfor HNPCC development starts with a muta-tion in the MMR genes followed by anothermutation in a gene such as APC. These twoevents lead to cellular hyperproliferation.Next, a mutation occurs leading to the inacti-vation of the wild-type allele of the MMR gene.Because of this MMR defect, mutations inother genes involved in tumor progression,such as deleted in colon cancer (DCC), p53,and K-ras, occur.

A variety of genes are responsible for thedifferent inherited forms of GI cancers. For

example, individuals with FAP, bearing germ-line mutations/deletions in the APC tumorsuppressor gene, account for only a small frac-tion of colon cancers in the United States(�1%). However, the majority of sporadic co-lon adenomas have also been found to containsingle allele alterations in APC and exhibit al-tered signaling of �-catenin, a protein nega-tively regulated by APC. Altered �-catenin sig-naling is inferred from immunohistochemicalstudies demonstrating that �-catenin is trans-located to the nucleus in the majority of epi-thelial cells in adenomas, whereas �-catenin isgenerally seen associated with the cell mem-brane in normal colonic epithelia. These datasuggest that the process of adenoma develop-ment selects for alterations in APC.

5 INTERVENTIONS

5.1 Prevention Strategies

Numerous investigators are taking advantageof our current knowledge of the mechanismsof carcinogenesis in human epithelial tissuesto develop strategies for disrupting this pro-cess and thereby preventing cancer. As dis-cussed earlier in this chapter, carcinogenesisproceeds by a multistep process, in which nor-mal epithelial tissues acquire aberrant growthproperties. These neoplastic cells progress tobecome invasive cancer. Historically, cancertherapy has addressed only the last phase ofthis process. Prevention strategies are now fo-cusing on pre-invasive, yet neoplastic lesions.

Prevention strategies generally influenceone or more of five processes in carcinogenesis(99). One strategy has been to inhibit carcino-gen-induced initiation events, which lead toDNA damage. An important caveat to thisstrategy is that the intervention must bepresent at the time of carcinogen exposure tobe effective. Once irreversible DNA damagehas occurred, this type of strategy is ineffec-tive in preventing cancer development.

Another strategy has been to inhibit initi-ated cell proliferation associated with the pro-motion stage of carcinogenesis. An advantageto this type of strategy is that interventionsaffecting promotion are effective after initiat-ing events have occurred. Because humansare exposed to carcinogenic agents (e.g., chem-

Molecular Biology of Cancer32

Page 33: Molecular Biology of Cancer - UC · PDF fileDepartments of Radiation Oncology/Cancer Biology Section Molecular and Cellular Biology ... 2 Molecular Biology of Cancer. are genetic mutations

icals in tobacco smoke, automobile exhaust)throughout their lifetimes, cancer preventiveagents that work after initiating events haveoccurred are desirable. Two strategies of de-creasing cell proliferation are induction ofapoptosis, or cell death, and differentiation,which may or may not be associated with apo-ptosis. Induction of either differentiation orapoptosis will stabilize or decrease, respec-tively, overall cell number in a tissue.

A final strategy for preventing cancer is toinhibit development of the invasive phenotypein benign, or non-invasive, precancers that oc-cur during the process of epithelial carcino-genesis.

Investigators are beginning to address thepossibility that the efficacy of cancer preven-tion strategies may depend on both geneticand environmental risk factors affecting spe-cific individuals. Mutations/deletion of theAPC tumor suppressor gene, discussed earlier,causes intestinal tumor formation in both ro-dents and humans. Increasing levels of dietaryfat increases intestinal tumor number in ro-dent models (100). However, mice with a de-fective APC gene develop tumors even on low-fat diets. Thus, dietary modifications mayreduce carcinogenesis in individuals without,but may be ineffective in individuals with, cer-tain genetic risk factors for specific cancers.Recently, several large randomized studiesconducted in the United States have failed todetect any protective effect of dietary fiber in-crease or dietary fat decrease on colon polyprecurrence (101).

5.2 Targets

Targets for cancer prevention strategies canbe either biochemical species produced by theaction of a physical or chemical carcinogen oran enzyme/protein aberrantly expressed as aconsequence of a genetic or environmentalrisk factor (the latter would include exposureto environmental carcinogens). In developingmechanism-based prevention or treatmentstrategies based on specific “targets,” it is cru-cial to establish that the “target” is present inthe target tissue (or cells influencing targettissue behaviors), causatively involved in thedisease process in question and modulated bythe intervention.

5.2.1 Biochemical Targets. One example ofa biochemical target produced by carcinogensis reactive oxygen species (ROS). Ionizing ra-diation is a complete carcinogen and producesmuch of its DNA damage through ROS (102).Several strategies for preventing ROS-in-duced cell damage have been developed. Theaminothiol, amifostine, inhibits radiation-in-duced DNA damage to a large degree by scav-enging free radicals produced by ionizing radi-ation. Amifostine and its derivatives suppressionizing radiation-induced transformationand carcinogenesis. Antioxidants, includingprotein and non-protein sulfhydrals and cer-tain vitamins, are effective modulators of ROSproduced by physical and chemical carcino-gens (103). Antioxidants are effective in inhib-iting carcinogenesis in some experimentalmodels, but their roles in human cancer pre-vention remains unclear. At least some agentswith antioxidant activity may increase carci-nogenesis in some tissues. Heavy smokers re-ceiving combinations of beta-carotene and vi-tamin A had excess lung cancer incidence andmortality, compared with control groups notreceiving this intervention (104).

Other examples of biochemical targets arethe dihydroxy bile acids, which are tumor pro-moters of colon cancer (105). Both genetic anddietary factors are known to influence intesti-nal luminal levels of these steroid-like mole-cules, whose levels are associated with coloncancer risk. Calcium reduces intestinal lumi-nal bile acid levels by several possible mecha-nisms, and dietary calcium supplementation isassociated with a small (�25%), but statisti-cally significant, reduction in colon polyp re-currence (106). This result requires cautiousevaluation, however, as similar levels of cal-cium supplementation have been associatedwith increased risk of prostate cancer (107).This example and the result of the beta-caro-tene study mentioned above underscore thetissue-specific differences in carcinogenesisand the difficulties of applying common di-etary components (e.g., calcium, antioxidants)in cancer prevention strategies in humans.

5.2.2 Cyclooxygenase-2 and Cancer. Cy-clooxygenase (COX) enzymes catalyze prosta-glandins from arachidonic acid. Prostaglan-dins play a role in biological processes

5 Interventions 33

Page 34: Molecular Biology of Cancer - UC · PDF fileDepartments of Radiation Oncology/Cancer Biology Section Molecular and Cellular Biology ... 2 Molecular Biology of Cancer. are genetic mutations

including blood clotting, ovulation, bone me-tabolism, nerve growth and development, andimmune responses (108). There are two COXisoforms, COX-1 and COX-2. COX-1 is consti-tutively expressed in most cell types and isnecessary for homeostasis of colonic epithe-lium and platelet aggregation. COX-2, on theother hand, is inducible by a variety of stimuliincluding growth factors, stress conditions,and cytokines (Fig. 1.12).

Several studies have implicated COX-2 incarcinogenesis. COX-2 protein levels, andtherefore, prostaglandin production, are up-regulated in many tumor types, including pan-creatic, gastric, breast, skin, and colon can-cers. Several lines of evidence suggest thatoverexpression of COX-2 plays an importantrole in colonic polyp formation and cancer pro-gression. COX-2 modulates metastatic poten-tial by inducing MMPs, which can be directlyinhibited by COX-2 inhibitors. In addition,cells overexpressing COX-2 secrete increasedlevels of angiogenic factors like VEGF andbFGF. COX-2 not only aids in invasion butalso inhibits apoptosis by up-regulating Bcl-2.

COX-2 has come under intensive study as atarget for colon cancer prevention. Multiplestudies have illustrated that COX-2 selectiveinhibitors suppress tumorigenesis in multipleintestinal neoplasia (Min) mice. COX-2 inhib-itors also inhibit tumor cell growth in immu-nocompromised mice (109). The same phe-

nomena has been illustrated in humanchemoprevention trials. Recent studies havelinked prolonged use of nonsteroidal anti-in-flammatory drugs (NSAIDs) to decreased co-lon cancer risk and mortality. NSAIDs inhibitthe cyclooxygenase enzymes, and new COX-2selective agents are gaining popularity in thetreatment of inflammation. NSAIDs that in-hibit both COX-1 and COX-2 have been asso-ciated with reduced cancer risk in severallarge epidemiology studies. Whether inhibi-tion of COX-1 and/or COX-2 is the optimalstrategy for reducing risks of certain cancers isunknown.

Because COX-2 is induced in certain neo-plastic tissues, the molecular regulation of itsexpression is being studied in a variety of ex-perimental models. Human and rodent celllines expressing various levels of COX-2 arebeing studied for genetic modifications thatlead to the dysregulation of COX-2. COX-2regulation occurs both transcriptionally andtranslationally, and this regulation differs de-pending on the species studied and the muta-tional status of the cell lines.

Signaling pathways leading to modulationof COX-2 expression are also being investi-gated. Both oncogenes and tumor suppressorgenes have been shown to modulate COX-2 incell model systems. The activation of the H-rasand K-ras oncogenes leads to induction ofCOX-2 expression in colon cancer cells. This

Membrane-boundphospholipids Arachidonic acid

phospholipases

COX-1COX-2

PGG2

PGH2

prostaglandins

prostacyclins

thromboxanes

cyclooxygenaseactivity

peroxidaseactivity

Figure 1.12. Cyclooxygenases catalyze prostaglandins from arachidonic acid. COX-2 is inducible bya variety of stimuli including growth factors, cytokines, and tumor promoters. PGH2 forms threeclasses of eicosanoids: prostaglandins, prostacyclins, and thromboxanes. Adapted from C. S. Williamsand R. N. DuBois, Am. J. Physiol., 270, G393 (1996).

Molecular Biology of Cancer34

Page 35: Molecular Biology of Cancer - UC · PDF fileDepartments of Radiation Oncology/Cancer Biology Section Molecular and Cellular Biology ... 2 Molecular Biology of Cancer. are genetic mutations

induction is mediated by the stabilization ofCOX-2 mRNA. Wild-type, full-length APCsuppresses COX-2 expression, suggesting thatnormal activity of this tumor suppressor genemay prevent cancer by inhibiting expressionof cancer-promoting genes like COX-2. APCdown-regulates COX-2 protein without affect-ing COX-2 mRNA levels. Thus, both ras andAPC regulate COX-2 expression by post-tran-scriptional mechanisms. TGF-�1 is anothertumor suppressor gene that influences expres-sion of COX-2. TGF-�1-mediated transforma-tion of rodent intestinal epithelial cells causesa significant induction of COX-2 protein ex-pression. TGF-�1 synergistically enhancesras-induced COX-2 expression by stabilizingCOX-2 mRNA. COX-2 expression is also influ-enced by the PI-3K pathway. Pharmacologicalinhibition of PI-3K or downstream PKB/Akt,as well as dominant-negative forms of Akt dra-matically reduce COX-2 protein levels.

5.2.3 Other Targets. Technologies such asDNA microarrays are identifying genes thatare aberrantly up-regulated in human intra-epithelial neoplasia (IEN). As discussed ear-lier, ODC, the first enzyme in polyamine syn-thesis, is up-regulated in a variety of IEN as aconsequence of specific genetic alterations. Di-fluoromethylornithine (DFMO), an enzymeactivated irreversible inhibitor of ODC, is apotent suppressor of several experimentalmodels of epithelial carcinogenesis and is be-ing evaluated in human cancer prevention tri-als (110). Pathways signaling cell behaviorsare also activated in specific cancers. A num-ber of agents, including NSAIDs and compo-nents of green and black teas, have beenshown to inhibit certain signaling pathways incell-type and tissue-specific manners.

5.3 Therapy

5.3.1 Importance of Studying Gene Expres-sion. Cancer, among other diseases, is causedby the deregulation of gene expression. Somegenes are overexpressed, producing abundantsupplies of their gene products, whereas othercrucial genes are suppressed or even deleted.The expression levels of genes associated withcancer influence processes such as cell prolif-eration, apoptosis, and invasion. Genes in-

volved in growth, for example, are often over-expressed in tumor tissues compared withnormal adjacent tissue from the same organ.It is imperative to elucidate which genes areoverexpressed or down-regulated in tumorsbecause these genes represent critical thera-peutic targets.

Researchers today generally concentrateon a few particular genes and study their reg-ulation, expression, and downstream signal-ing using conventional molecular biologytools. With the onslaught of new genome data,and the development of the GeneChip, scien-tists are now able to study the expression lev-els of numerous genes simultaneously. Theability to analyze global profiles of gene ex-pression in normal tissue compared withtumor tissue can help reveal how gene expres-sion affects the overall process of carcinogen-esis.

5.3.2 cDNA Microarray Technology.cDNA microarray technology is based on thesimple concept of DNA base pairing. cDNAfrom tumor samples hybridize with the com-plementary DNA sequences on the chip. TheDNA sequences are the target genes that willbe studied for expression levels in particulartissues. These sequences, or probes, can be inthe form of known oligos, DNA encoding thefull-length gene, open reading frames (ORFs),or sometimes even the entire genome of anorganism like Saccharomyces cerevisiae.Genes can be chosen by their proximity toeach other on a chromosome or their similarfunctions. cDNA probes are then spotted ontoa glass slide or computer chip (GeneChip), us-ing a variety of different robotic techniques. Atypical microarray slide will contain approxi-mately 5000 genes.

cDNA microarray is particularly useful tothe field of cancer biology because it allowsscientists to study changes in gene expressioncaused when a normal tissue becomes neoplas-tic. In addition, normal tissue can be com-pared with preneoplastic lesions as well asmetastatic cancer, to fully examine the entiretumorigenic process. The mRNA is extractedfrom cell lines or tissue and is reverse tran-scribed into the more stable form of cDNA.The cDNA is then labeled with reporters con-taining two colored dyes, rhodamine red, Cy3,

5 Interventions 35

Page 36: Molecular Biology of Cancer - UC · PDF fileDepartments of Radiation Oncology/Cancer Biology Section Molecular and Cellular Biology ... 2 Molecular Biology of Cancer. are genetic mutations

and fluorescien green, Cy5. The cDNA is thenhybridized to the DNA on the microarrayslide. The slides are exposed to a laser beam,causing the dyes to give off their respectiveemissions and the relative expression levels ofthat gene are read and processed.

A similar technique to cDNA microarraythat allows for multigene expression analysisis serial analysis of gene expression (SAGE)(Fig. 1.13). SAGE is based on the principlethat a 9–10 nucleotide sequence contains suf-ficient information to identify a gene. Theseshort nucleotide sequences are amplified bypolymerase chain reaction (PCR) and then

30–50 of these SAGE “tags” are linked to-gether as a single DNA molecule. These longDNA molecules are sequenced and the num-ber of times that a single “tag” appears corre-lates to that gene’s expression level. Proof ofconcept for this technique was illustrated in astudy of gene expression in pancreatic cells.The most abundant “tags” found were thosethat encoded highly expressed pancreatic en-zymes like trypsinogen 2. cDNA microarraymethodology has also been validated by stud-ies showing that expression data for tumor celllines grown in tissue culture conditions can beclassified according to their tissue of origin.

Lymphocyte mRNAfrom healthy person

Lymphocyte mRNAfrom affected patient

Lymphocyte mRNAfrom healthy person

Isolate 9 base pair "tag" froma defined region of the cDNAs

Sequence and count number of each tag

Link tags

Lymphocyte mRNAfrom affected patient

AAAAAAAAAAAAAAAAAAAAAAAA

Single-stranded cDNA labeledwith two fluorescent dyes

Add equal amounts to microarrayof cDNAs from human lymphocytes

Incubate and scan

or

0

Genes (Healthy, black: Sick, white)

10

Exp

ress

ion

leve

l

Figure 1.13. Comparison of cDNA microarray and SAGE technologies. At left is a diagram of themicroarray assay for gene expression; the SAGE technique is illustrated at right. Here, the proce-dures assess how gene expression differs in lymphocytes from a healthy person and those from aperson fighting off an infection. Reprinted with permission from K. Sutliff, Science, 270, 368 (1995).American Association for the Advancement of Science.

Molecular Biology of Cancer36

Page 37: Molecular Biology of Cancer - UC · PDF fileDepartments of Radiation Oncology/Cancer Biology Section Molecular and Cellular Biology ... 2 Molecular Biology of Cancer. are genetic mutations

5.3.3 Discoveries from cDNA MicroarrayData. The contribution of microarray tech-nology is influential in both the basic under-standing of cancer pathology as well as in drugdiscovery and development. These studies re-veal genes that may prove to be important di-agnostic or prognostic markers of disease.They also can be used to predict adverse reac-tions to chemotherapies if mRNA from drug-treated cells is hybridized to panels of genesrelated to liver toxicity or the immune re-sponse.

Microarray technology also corroboratesmany in vitro cell studies that are criticized forignoring the important role of other cell typesin the tumor microenvironment. This technol-ogy can aid in distinguishing between celltype–specific or tumor-specific gene expres-sion. For example, SAGE analysis of colon tu-mors and colon cancer cell lines showed 72% ofthe transcripts expressed at reduced levels incolon tumors were also expressed at reducedlevels in the cell lines. One interesting findingfrom this study was that two commonly mu-tated oncogenes, c-fos and c-erbB3, werefound to be expressed at higher levels in nor-mal colonic epithelium than in colonic tumors;this contradicts reports that these oncogenesare up-regulated in transformed cells com-pared with normal cells. Again, microarrayanalysis is helping to merge cell biology stud-ies with whole tumor biology.

Activation of the c-myc oncogene is a com-mon genetic alteration occurring in many can-cers. A cDNA microarray study found that c-myc activation leads to down-regulation ofgenes encoding extracellular matrix proteins,and thus, may play a role in regulating celladhesion and structure. C-myc has also beenassociated with cell proliferation, which wasillustrated by up-regulation of the geneseIF-5A and ODC. Another study of colon tu-mors revealed that only 1.8% of the 6000 tran-scripts studied were differentially expressedin normal tissues and tumors (111). Studiessuch as these suggest the critical importanceof these differentially regulated genes in thecancer phenotype.

In addition to oncogene activation, the ef-fects of tumor suppressor genes have been in-vestigated through microarray technology.Over 30 novel transcripts were identified as

regulated by p53 induction (112). Such a greatnumber of genes simultaneously linked to p53expression would not have been possible with-out SAGE technology. However, only 8% ofthese new genes were induced in normal cellscompared with p53 knockout cells, suggestingthat most of these p53-dependent genes arealso dependent on other transcription factors.This is just one example of how microarraytechnology may be able to look at crosstalk insignaling pathways.

5.3.4 Limitations of Microarray Technolo-gies. Although cDNA microarray and SAGEtechnologies are quickly identifying new genesinvolved in tumorigenesis, there are signifi-cant limitations to these strategies. First, theexpression pattern of a gene only provides in-direct information about its function; a newgene may be classified as necessary for a cer-tain biological process, but its exact role inthat process cannot be determined. Second,mRNA levels do not always correlate with pro-tein levels, and even protein expression maynot translate into a physiological effect. Third,the up-regulation or suppression of a genemay be either the cause or the effect of a dis-ease state and microarray technology does notdistinguish between the two possibilities.

Both cDNA microarray and SAGE analysesrequire verification of changes in gene expres-sion by Northern blots. Modest changes ingene expression are often overlooked whendata is reported in terms of fourfold or greaterchanges. Because the ability to detect differ-ences in gene expression is dependent on themagnitude of variance, a small induction orsuppression of a gene may be discarded as in-consequential when it may actually be criticalfor downstream signaling of other genes.

5.4 Modifying Cell Adhesion

5.4.1 MMP Inhibitors. Several MMP inhib-itors are currently being developed for cancertreatment. If MMPs do play an integral role inmalignant progression, then pharmacologicalinhibition of MMPs could inhibit tumor inva-siveness. The inhibition of MMP function iscurrently the focus of most antimetastatic ef-forts. MMP inhibitors fall into three catego-ries: (1) collagen peptidomimetics and non-

5 Interventions 37

Page 38: Molecular Biology of Cancer - UC · PDF fileDepartments of Radiation Oncology/Cancer Biology Section Molecular and Cellular Biology ... 2 Molecular Biology of Cancer. are genetic mutations

peptidomimetics, (2) tetracycline derivatives,and (3) bisphosphonates. The peptidomimeticMMP inhibitors have a structure that mimicsthat of collagen at the site where the MMPbinds to it. Batimastat, a peptidomimetic in-hibitor, was the first MMP inhibitor to be eval-uated in cancer patients and is not orally avail-able. Matimastat is orally available and iscurrently in phase II and III clinical trials(113). When bound to the MMP, these inhibi-tors chelate the zinc atom in the enzyme’s ac-tive site. There are several nonpeptidomi-metic inhibitors that are also in variousphases of clinical trials. These are more spe-cific than their peptidic counterparts and haveexhibited antitumor activity in preclinicalstudies (113).

Tetracycline derivatives inhibit both theactivity of the MMPs and their production.They can inhibit MMP-1, -3, and -13 (the col-lagenases) and MMP-2 and -9 (the gelatinases)by several different mechanisms. These mech-anisms include (1) blocking MMP activity bychelation of zinc at the enzyme active site, (2)inhibiting the proteolytic activation of the pro-MMP, (3) decreasing the expression of theMMPs, and (4) preventing proteolytic and ox-idative degradation of the MMPs.

The mechanism of action of the bisphos-phonates has not been elucidated, but theyhave been used extensively for disorders incalcium homeostasis and recently in breastcancer and multiple myeloma patients to pre-vent bone metastases (114). Clodronate, abisphosphonate, inhibited expression of MT1-MMP RNA and protein in a fibrosarcoma cellline and effectively reduced the invasion ofmelanoma and fibrosarcoma cell lines throughartificial basement membranes (115).

5.4.2 Anticoagulants. One theory sur-rounding the invasion process is that blood-clotting components may play a role in metas-tasis by either trapping the tumor cells incapillaries or by facilitating their adherence tocapillary walls. Large numbers of tumor cellsare released into the bloodstream duringthe metastatic process, and they must be ableto survive the wide range of host defensemechanisms. Tumor cells have been shown tointeract with platelets, lymphocytes, and leu-

kocytes, and this may serve to promote metas-tasis. Studies have been done that inhibit tu-mor cell-platelet interactions, and these haveresulted in a decreased probability of metasta-sis formation. It has also been shown that fi-brin is always located in and around cancerouslesions, which may indicate that the cells usethe fibrin structure as a support on which toattach themselves and grow. It may also serveas protection against host inflammatory cellsso that the tumor is not destroyed.

Treating hepatic metastases of a humanpancreatic cancer in a nude (lacking a thymus)mouse with prostacyclin, a potent inhibitor ofplatelet aggregation, led to a significant reduc-tion in the mean surface area of the liver cov-ered with tumor compared with the untreatedcontrol group (116). Many other groups havereported a reduction in metastatic potentialwith treatment of prostacyclin and prostacy-clin-analogues, such as iloprost and cicaprost.There are currently over 50 different clinicaltrials in varying phases underway to deter-mine the efficacy of these anticoagulant ther-apies. Most of these trials are in combinationwith other conventional anti-cancer regimens.So far, the experimental evidence indicatesthat anticoagulants or inhibitors of plateletaggregation are useful in the prevention ofmetastases.

5.4.3 Inhibitors of Angiogenesis. The growthand expansion of tumors and their metastasesare dependent on angiogenesis, or new bloodvessel formation. Angiogenesis is regulated bya complex of stimulators and inhibitors (Fig.1.14). The balance between the positive andnegative regulators of angiogenesis inside atumor environment is important for the ho-meostasis of microvessels. Tumor cells can se-crete proangiogenic paracrine factors, whichstimulate endothelial cells to form new bloodvessels. The use of angiogenesis inhibitorsmay be a potential mode of therapy and is stillin early clinical trials. This type of therapywould be a way of controlling the diseaserather than eliminating it. Whereas toxicitymay not be a major problem, adverse effectsmay be expected in fertility and wound heal-ing.

Molecular Biology of Cancer38

Page 39: Molecular Biology of Cancer - UC · PDF fileDepartments of Radiation Oncology/Cancer Biology Section Molecular and Cellular Biology ... 2 Molecular Biology of Cancer. are genetic mutations

5.5 Prospects for Gene Therapy of Cancer

Gene therapy is the transfer of genetic mate-rial into cells for therapeutic purpose. Genetransfer technology has become available afterextensive study of molecular mechanisms ofmany diseases and improvement of tech-niques for manipulating genetic materials inthe laboratory. Concepts for genetic therapy ofcancer were developed based on knowledgethat neoplasia is a molecular disorder result-ing from loss of expression of recessive tumorsuppressor genes and activation of dominantoncogenes.

Cancer gene therapy is aimed at correctinggenetic mutations found in malignant cells ordelivering biologically active material againstcancer cells. One approach used in gene ther-apy of cancer is gene replacement/correctionto restore the function of a defective homolo-gous gene or to down-regulate oncogenic ex-pression in somatic cells. Another approach isimmune modulation by introduction of thera-peutic genes, such as cytokines, into the targetcells to treat cancer by stimulating an immuneresponse against the tumor. Molecular ther-apy by activating prodrugs (e.g., ganciclovir,5-fluorocytosine) within tumor cells and sui-cide gene therapy approaches have alreadybeen successful in early clinical trials. Thehigh performance of these approaches fullydepends on the efficacy and specificity of ther-apeutic gene expressing and delivery systems.

5.5.1 Gene Delivery Systems. The exoge-nous genetic material (the transgene) is usu-ally introduced into tumor cells by a vector. Avector, or plasmid, is a circular DNA sequence

that is designed to replicate inserted foreignDNA for the purpose of producing more pro-tein product. Plasmids designed for gene ther-apy applications usually contain the gene ofinterest and regulatory elements that enhancethe gene’s expression. The ideal vector forgene therapy is one that would be safe, havehigh transfection efficiency, and be easy to ma-nipulate and produce in large quantities. Itwould be efficient at delivering genetic mate-rial and selectively transducing cells within atumor mass. The vector would be immuno-genic for the recipient and would express thegene in a regulated fashion and at high levelsas long as required.

There are two main approaches for the in-sertion of gene expressing systems into cells.In the ex vivo technique, cells affected by thedisease are transfected with a therapeuticgene in vitro for the expression of exogenousgenetic material. After viral propagation, rep-lication is rendered incompetent and thesecells can be transplanted into the recipient. Inthe in vivo technique, vectors are inserted di-rectly into target tissue by systemic injectionsof the gene expressing system.

The simplest delivery system is a plasmidby itself, or so-called naked DNA. Direct injec-tions of DNA have been successfully used totransfect tissues with low levels of nucleaseactivity in muscle tissue (117), liver (118), andexperimental melanoma (119). Systemic injec-tion of naked DNA is, in general, much lessefficient because serum nucleases degradeplasmid DNA in the blood within minutes(120).

Stimulators Inhibitors

• Fibroblast growth factor

• VEGF

• HGF

• PDGF

• EGF

• Angiostatin

• Endostatin

• Thrombospondin-1

• Troponin I

• Vasostatin

Stimulation of angiogenesisand neovascularization

Inhibition of angiogenesisand vascular quiescence

Figure 1.14. Stimulators and inhibitorsof angiogenesis. Under physiological con-ditions, the balance of factors that affectangiogenesis is precisely regulated. How-ever, under pathophysiological condi-tions, normal angiogenesis is disturbedbecause of the continued production ofstimulators.

5 Interventions 39

Page 40: Molecular Biology of Cancer - UC · PDF fileDepartments of Radiation Oncology/Cancer Biology Section Molecular and Cellular Biology ... 2 Molecular Biology of Cancer. are genetic mutations

To protect DNA on systemic application, itis usually complexed with viruses or with cat-ionic lipids, polymers, or peptides. The result-ing complex protects the DNA from the attackof nucleases and potentially improves trans-fection efficiency and specificity on multiplelevels through interaction of DNA complexeswith the various biological barriers.

The choice of viral or non-viral (synthetic)delivery strategy depends on localization andtype of affected tissue, as well as on therapeu-tic approach. Viral vectors use the ability ofviruses to overcome the cellular barriers andintroduce genetic material either through theintegration of the vector into the host genome(retroviruses, lentiviruses, adeno-associated vi-ruses) or by episomal delivery (adenoviruses)followed by stable gene expression (Fig. 1.15).

5.5.1.1 Viral Vectors. Retroviral vectorshave been used for ex vivo gene delivery andare the most useful vectors for stably integrat-ing foreign DNA into target cells. Retrovirusesare enveloped viruses that contain 7- to 12-kbRNA genomes. After the virus enters the cellsthrough specific cell surface receptors, its ge-nome is reverse transcribed into double-stranded DNA and subsequently integratedinto the host chromosome in the form of a pro-virus. The provirus replicates along with thehost chromosome and is transmitted to all ofthe host cell progeny. Because the retrovirusgenome is relatively small and well character-ized, it was possible to engineer a vector en-coding only the transgene without replicationcompetent viruses (RCV) or virus structuralgenes.

R

L

Retrovirus

Lentivirus

A Adenovirus

V

DNA

Lipoplex

Vaccinia virus

Plasmid DNA

Plasmid DNA+cationic liposome

RL A

R LA

V

V

DNADNA

DNA

Lipoplex

DNA

A

Integrating vectors

Nonintegrating vectors

AAV Adeno-associated virus

AAV

AAV

R--L

AAV-

Figure 1.15. Virus particles bind to specific receptors on the surface of target cells. These vectors areinternalized and their genome enters the cells. In the case of retroviruses, the single-stranded RNAgenome is converted into double-stranded DNA by the reverse transcriptase enzyme encoded by thevirus. The double-stranded DNA is taken up by the nucleus and integrated within the host genome asa provirus. The integration is random for retroviruses. Lentiviruses have a similar life cycle. Adeno-virus binds to specific receptors on the surface of susceptible cells and are then absorbed and inter-nalized by receptor-mediated endocytosis. The viral genome enters the cytoplasm of the cell and thedouble-stranded DNA genome is taken up by the nucleus. Vaccinia virus replicates in the cytoplasmof cells. DNA delivered by lipoplex and other nonviral systems enters cells through electrostaticinteractions (endocytosis, phagocytosis, pinocytosis, and direct fusion with cell membrane). DNA isreleased before entry into the nucleus, where it stays as an episome.

Molecular Biology of Cancer40

Page 41: Molecular Biology of Cancer - UC · PDF fileDepartments of Radiation Oncology/Cancer Biology Section Molecular and Cellular Biology ... 2 Molecular Biology of Cancer. are genetic mutations

The most widely used retrovirus vectorsare based on murine leukemia viruses (MLV).The lack of specificity of these vectors is a ma-jor obstacle for appropriate and controlled ex-pression of foreign genes. Retroviruses are notefficient for direct in vivo injection because ofinactivation by the host immune system (121).To circumvent this, cis-acting viral sequences,such as long terminal repeats (LTRs), transferRNA (tRNA) primer binding sites, and polypu-rine tracts, have been used for developingpackaging systems of retrovirus vectors. Manyrecombinant retrovirus vectors are designedto express two genes, one of which is often aselectable marker. New strategies for expres-sion, such as splicing, transcription from het-erologous promoters, and translation directedby an internal ribosome entry signal (IRES),have been used for expression of the secondgene. Attempts have also been made toachieve efficient gene delivery by targetingretroviral integration through modifying pro-tein sequences in the viral envelope (122).These modifications include various targetingligands, particularly ligands for the humanEGFR, erythropoetin receptor, and singlechain antibody fragments against the low-density lipoprotein (LDL) receptor.

Examples of lentiviruses are the humanimmunodeficiency virus (HIV-1), the equineinfectious anemia virus, and the feline immu-nodeficiency virus (123). Although lentivi-ruses also have an RNA genome, their advan-tage compared with other retroviruses is theability to infect and stably integrate into non-dividing cells. To create a safe gene transfervector based on the HIV-1 genome, the ge-nome was altered and mutated to produce rep-lication-defective particles. Several studies,both in vitro and in vivo, have shown success-ful gene transfer, including transduction ofnon-dividing hematopoetic cells at high effi-ciencies (up to 90%) and stable gene expres-sion in several target tissues of interest suchas liver (8 weeks) and muscle and brain (6months) with no detectable immune response(124). At the same time, the safety concernstill remains for in vivo applications of thisvector.

Vaccinia virus is a member of the Poxviri-dae family, which possesses a complex DNAgenome encoding more than 200 proteins. The

advantages of using vaccinia viruses for genetransfer include their ability to accommodatelarge or multiple gene inserts, to infect cellsduring different stages of the cell cycle, andtheir unique feature to replicate in the cyto-plasm. Recombinant vaccinia vectors can beconstructed using homologous recombinationafter transfection of vaccinia virus-infectedcells with plasmid DNA constructs. This vec-tor has been used in clinical trials to delivergenes encoding tumor antigens such as mela-noma antigen (MAGE-1), carcinoembryonicantigen (CEA), prostate-specific antigen(PSA), interleukins (e.g., IL-1�, IL-12), andcostimulatory molecule B7 (125).

In recent years, there has been a great in-terest in the use of adenoviral vectors for can-cer gene therapy. The main reasons for thisare the ease in construction of adenoviruses inthe laboratory and their ability to grow to hightiters, infect a variety of cell types, and pro-duce the heterologous protein of interest individing and non-dividing cells. Adenovirusesare also characterized by efficient receptor-mediated endocytosis, mediated by its fiberprotein, and on infection of cancer cells, theyexhibit high levels of transgene expression(126). They often are used to transfer genes oflarge sizes because of their high packaging ca-pacity (up to 36 kb). Adenoviral vectors do notintegrate into the host chromosomes, andtherefore, they are degraded by the host. Thisresults in a short-term expression of the trans-duced gene, which, nevertheless, could be suf-ficient to achieve the cancer gene therapy effi-cacy. Adenoviruses are widely used for directin vivo injections. Adenoviruses are DNA-con-taining, non-enveloped viruses.

The two most commonly used adenovirusesfor recombinant vectors are Ad2 and Ad5,mainly because their genomes have been bestcharacterized and because these viruses havenever been shown to induce tumors. Adenovi-ruses, like other viral vectors, lack cell andtissue specificity. To improve targeted genedelivery, attempts have been made to coupleligands or antibodies to the adenovirus capsidproteins (127). Specificity of the transgene ex-pression also can be introduced by using tissuespecific antigens, such as CEA for the treat-ment of pancreatic and colon cancers, mucin(MUC-1) promoters for breast cancer cells, al-

5 Interventions 41

Page 42: Molecular Biology of Cancer - UC · PDF fileDepartments of Radiation Oncology/Cancer Biology Section Molecular and Cellular Biology ... 2 Molecular Biology of Cancer. are genetic mutations

pha-fetoprotein promoters for hepatocellularcarcinoma, and the tyrosinase promoter formelanoma (126). In vivo administration of ad-enoviral vector has been extensively used inpreclinical and clinical cancer therapy (128).

There are many regulatory elements con-trolling cell type–specific gene expression andinducible sequences within promoters thathave been used in construction of viral vectorsfor cancer therapy. Vector systems that in-clude cell type–specific promoters or elementsresponding to regulatory signals represent away for a safe, selective, and controlled expres-sion of therapeutic genes that could increaseefficacy and stability of gene expression (Table1.5).

Vectors based on adenoassociated viruses(AAV) also have been successfully used totransfer genes. AAV is a small, single-stranded DNA virus that requires a helper vi-rus for infection, usually an adenovirus or her-pesvirus. AAV vectors can be used for thedelivery of antisense genes, “suicide” genetherapy, and recently, for the delivery of anti-angiogenic factors. Recent studies in the areaof vector design have been focused on condi-tional expression that can be induced by anti-biotics (129), heat shock (130), or other smallmolecules (131).

5.5.1.2 Non-Viral Gene Delivery Systems.Non-viral gene delivery systems are based onnon-covalent bonds between cationic carriermolecules (e.g., lipids or polymers) and thenegatively charged plasmid DNA. Complexesof DNA with three main groups of materials,i.e., cationic lipids (lipoplex) such as CTABand DMRI, polymers (polyplex) such as poly-L-lysine and polyathylenimine, or peptideshave been evaluated as synthetic gene deliverysystems (132). The formation of these com-plexes, which is generally based on electro-static interactions with the plasmid DNA, isdifficult to control as they depend on both thestoichiometry of DNA and complexing agentand on kinetic parameters (e.g., speed of mix-ing and volumes). It has been shown that DNAis efficiently condensed and protected fromnucleases at higher lipid:DNA ratios, provingthat the positive charges of the complexes areimportant for the interaction with cells invitro and in vivo. Although the resulting par-ticles are stable, they have a high tendency tointeract non-specifically with biological sur-faces and molecules.

Lipoplexes are actively used in clinical tri-als for in vivo and ex vivo delivery of genesencoding cytokines, immunostimulatory mol-ecules, and adenoviral genes (133). In vivo,

Table 1.5 Cell-Type Specific Promoters for Targeted Gene Expression

Promoter Target Cell/Tissue Therapeutic Gene

PEPCK promoter Hepatocytes Neomycin phosphotransferase,Growth hormone

AFP promoter Hepatocellular carcinoma HSV-tk, VZV-tkMMTV-LTR Mammary carcinoma TNF�WAP promoter Mammary carcinoma Recombinant protein C�-casein Mammary carcinoma In developmentCEA promoter Colon and lung carcinoma HSV-tk, CDSLPI promoter Carcinomas HSV-tk, CDTyrosinase promoter Melanomas HSV-tk, IL-2c-erbB2 promoter Breast, pancreatic, gastric

carcinomasCD, HSV-tk

Myc-max-responsive element Lung HSV-tk

Therapy-Inducible Tissues

Egr-1 promoter Irradiated tumors TNF�Grp78 promoter Anoxic, acidic tumor tissue Neomycin phosphotransferaseMDR1 promoter Chemotherapy-treated tumors TNF�HSP70 Hyperthermy-treated tumors IL-2

Molecular Biology of Cancer42

Page 43: Molecular Biology of Cancer - UC · PDF fileDepartments of Radiation Oncology/Cancer Biology Section Molecular and Cellular Biology ... 2 Molecular Biology of Cancer. are genetic mutations

these interactions may compromise the tissue-specific delivery of the complexes, creating un-even biodistribution and transgene expressionin the body, particularly, in lungs. To over-come this problem, the complexes can be in-jected either into the vasculature or directlyinto the affected organ (134).

The combinatorial gene delivery approachuses the whole virus, either replication defi-cient or inactivated, or only essential viralcomponents, together with the non-viral sys-tem. Systems, based on adenovirus (“adeno-fection”), or viral proteins that are required totrigger efficient endosomal escape, and poly-plex and lipoplex non-viral systems haveshown improvement in transfection efficiencyand resistance to endosomal degradation(135).

5.6 Gene Therapy Approaches

5.6.1 Immunomodulation. This approachemploys the patient’s physiological immuneresponse cascade to amplify therapeutic ef-fects (136). Most patients with cancer lack aneffective immune response to their tumors.This could be caused by defects in antigen pre-sentation, stimulation, or differentiation ofactivated T cells into functional effector cells.Antitumor immunity response requires par-ticipation of different immune cells, includinghelper effector T-cells (Th), cytotoxic T-lym-phocytes (CTLs), and natural killer (NK) cells.Activation of CD4� and CD8� T-cells requiresat least two major signals. The first signal istriggered by binding of complexes of T-cell re-ceptor (TCR) and specific antigenic peptidewith MHC-class II or I molecules, respectively.The second signal for CD4� T-cells is providedby engagement of CD28 on the T-cell surfaceby members of the B7 family of costimulatorymolecules on the surface of professional anti-gen-presenting cells. The nature of second sig-nal for CD8� T-cells has not been completelyunderstood but requires the presence ofhelper CD4� T-cells. Following activation andclonal expansion, activated CD4� T-cells dif-ferentiate into helper effector cells of eitherthe Th1 or Th2 phenotype. Th1 cells producecytokines, such as IL-2, interferon-�, andTNF, that stimulate monocytes and NK cells

and promote the differentiation of activatedCD8� T-cells into CTLs.

The growing understanding of the biologi-cal basis of antigen-specific cellular recogni-tion and experimental studies of an antitumoreffect mediated through the cellular immunesystem helped to develop various immuno-modulation strategies. Modulation of immuneresponse can be achieved through stimulationand modification of immune effector cells, en-abling them to recognize and reject cells thatcarry a tumor antigen. Additionally, tumorcells can be genetically modified to increaseimmunogenicity and trigger an immuneresponse.

Cytokine levels are relatively low in cancerpatients. To correct for this deficiency, cyto-kines can be introduced as recombinant mole-cules, and this is advantageous in controllingtheir blood concentration and biological activ-ity. Because cytokines are relatively unstablein vivo, cancer patients have to receive a largeamount of the recombinant protein to main-tain the required blood concentration for bio-logical activity. Administration of the proteinis often toxic to the patients. Another thera-peutic approach is the introduction of genesencoding various cytokines, costimulatorymolecules, allogenic antigens, and tumor-as-sociated antigens into tumors (137). Previouspreclinical studies have shown that cytokinesthat facilitate Th1 cell-mediated immune re-actions but not Th2 cell-mediated reactions,when produced in tumors, are effective for an-titumor responses. In addition, cytokines orcostimulatory molecules delivered to tumorcells may enhance the transfer of tumor anti-gens to antigen-presenting cells. The most po-tent known antigen-presenting cells for ac-tively stimulating specific cellular immuneresponses are dendritic cells. Ex vivo gene de-livery to cultured dendritic cells or direct invivo gene delivery to antigen-presenting cellscan be more efficient in stimulating cellularantitumor immunity (138).

Several technical problems of expressingsufficient amounts of immunostimulatoryproteins in appropriate target cells remain un-solved, but the potential of immune modula-tion gene therapy is high. Immunotherapy tri-als also contribute to the present knowledge of

5 Interventions 43

Page 44: Molecular Biology of Cancer - UC · PDF fileDepartments of Radiation Oncology/Cancer Biology Section Molecular and Cellular Biology ... 2 Molecular Biology of Cancer. are genetic mutations

how antitumor responses can be effectivelyproduced in cancer patients.

5.6.2 Suicidal Gene Approach. Elimina-tion of cancer cells can be accomplished by theintroduction of vectors that specifically ex-press death promoting genes in tumor cells.One method, called suicide gene therapy, in-volves the expression of a gene encoding anenzyme, normally not present in human cells,that converts a systemically delivered non-toxic prodrug into a toxic agent. The toxinshould kill the cancer cells expressing the geneas well as the surrounding cells not expressingthe gene (bystander effect).

The herpes simplex virus thymidine kinasetype 1 (HSV-tk) gene was initially used forlong-term replacement gene therapy becauseit is about 1000-fold more efficient than mam-malian thymidine kinase at phosphorylatingthe nontoxic prodrug ganciclovir (GCV) intoits toxic metabolite ganciclovir triphosphate.The efficacy of HSV-tk transduction of tumorsfollowed by ganciclovir therapy has been con-firmed by systemic administration of ganciclo-vir after intratumoral injection of fibroblaststransduced with an HSV-tk retroviral vectorin several preclinical models (139). The molec-ular mechanism of HSV-tk therapy is based oninduction of apoptosis in target cells throughaccumulation of p53 protein (Fig. 1.16). Clini-cal trials of HSV-tk suicide gene therapy,

where ganciclovir was given after the retroviralor adenoviral introduction of HSV-tk gene, havebeen conducted in patients with brain tumors,melanoma, or mesothelioma (140–142) (Fig.1.16).

Another suicide gene under active investi-gation for cancer therapy is the cytosinedeaminase (CD) gene. CD converts the non-toxic fluoropyrimidine 5-fluorocytosine to5-fluorouracil. Transduction of the CD ren-ders tumor cells sensitive to 5-fluorocytosinein vitro and in vivo. The CD/5-fluorocytosinesystem has been used in a clinical trial, whereadenovirus expressing the CD gene was in-jected intratumorally into hepatic metastasesfrom colorectal cancer (143). As with HSV-tkgene transfer, evidence exists that cytosine-deaminase gene transfer into tumor cells pro-motes antitumor immune responses. The ma-lignancies targeted with suicide gene therapyin the field of pediatric oncology are brain tu-mors, neuroblastoma, and acute lymphoblas-tic leukemia (144).

5.6.3 Targeting Loss of Tumor SuppressorFunction and Oncogene Overexpression. Sev-eral tumor suppressor genes, including p53,Rb, and APC, have been identified by theirassociation with hereditary cancers. Manysporadic tumors harbor inactivating or reces-sive mutations in one or more tumor suppres-sor genes. Gene transfer techniques can be ap-

Substrates

GCV-PPPCytochrome c

p53

CD95

FADD

Caspase-8Golgi

Caspases

Nucleus

Mitochondria

Figure 1.16. Mechanisms of thymidine ki-nase (TK) ganciclovir (GCV)-induced apo-ptosis. TK phosphorylates the nontoxic pro-drug GCV to GCV-triphosphate (GCV-PPP), which causes chain termination andsingle-strand breaks on incorporation intoDNA. TK/GCV induces p53 accumulation,which can cause translocation of preformeddeath receptor CD95 from the Golgi appara-tus to the cell surface without inducing denovo synthesis of CD95. The signaling com-plex then is formed by CD95, the adaptermolecule Fas-associated death domain(FADD) protein, and the initiator caspase-8,which leads to cleavage of caspases causingapoptosis. TK/GCV also leads to mitochon-dria damage, including loss of mitochon-drial membrane potential and the release ofcytochrome c inducing caspase activationand nuclear fragmentation.

Molecular Biology of Cancer44

Page 45: Molecular Biology of Cancer - UC · PDF fileDepartments of Radiation Oncology/Cancer Biology Section Molecular and Cellular Biology ... 2 Molecular Biology of Cancer. are genetic mutations

plied to introduce wild-type copies of tumorsuppressor genes into malignant cells, thuspotentially reversing the neoplastic pheno-type. The p53 tumor suppressor gene has beenof interest because p53 mutation occurs com-monly in a variety of human cancers, includ-ing breast, lung, colon, prostate, bladder, andcervix. The use of adenoviral vectors to deliverthe p53 transgene to human tumors is nowunder evaluation in several clinical trials(145). The overexpression of Fas ligand causedby adenovirus-mediated wild-type p53 genetransfer induces neutrophil infiltration intohuman colorectal tumors, which may play acritical role in the bystander effect of p53 genetherapy (146).

Besides the p53 gene, other tumor suppres-sor genes that regulate the cell cycle have beenused in cancer gene therapy. Among them areRb, BRCA1, PTEN, p16, E2F, and fragile his-tidine triad (FHIT) genes. Clinical trials withBRCA1 and Rb have been initiated (147).

Protooncogenes, in contrast to tumor sup-pressor genes, gain dominant mutation result-ing in excessive expression of their proteinproducts, which lead to development of themalignant phenotype. Three members of theRas family of oncogenes (H-ras, K-ras, and N-ras) are among the most commonly activatedoncogenes in human cancers. Several strate-gies have been designed to combat K-ras mu-tations, including antisense nucleotide, ri-bozymes (148–150), and intracellular single-chain antibodies (151). cDNA encodingantisense RNA can be delivered using the viralvector system approach. In vivo gene therapywith K-ras, c-fos, and c-myc antisense nucleo-tides is currently being applied in clinical tri-als.

5.6.4 Angiogenesis Control. Gene therapyoffers a new strategy for the delivery of angio-genesis inhibitors. By engineering and deliv-ering vectors that carry the coding sequencefor an antiangiogenic protein, it is possible toproduce high levels of antiangiogenic factorsin the tumor location or to systemically pre-vent the growth of distant metastasis. Severalangiogenic inhibitors, such as angiostatin(152), endostatin (153), plasminogen activatorinhibitor type 1 (154), and truncated VEGFreceptor (155), have been tested using this ap-

proach. These studies have demonstrated thatretroviral and adenoviral vectors could beused to inhibit endothelial cell growth in vitroand angiogenesis in vivo. The inhibition of tu-mor-associated angiogenesis results in in-creased apoptotic tumor cell death, leading toinhibition of tumor growth.

5.6.5 Matrix Metalloproteinase. As men-tioned earlier in the chapter, MMPs are capa-ble of proteolytic degradation of stromal ECM,which is essential in cancer cell migration andinvasion, as well as in tumor-induced angio-genesis. The activity of MMPs in vivo is inhib-ited by TIMPs, small secreted proteins withmolecular weight of between 20 and 30 kDa.TIMPs inhibit MMPs by binding to both thelatent and active forms of MMPs. The follow-ing properties of TIMPs such as secretion, dif-fusion (TIMP-1, -2 and -4), induction of apo-ptosis (TIMP-3), and inhibition of multipleMMPs make them very attractive tools forgene therapy application.

Inhibition of cancer cell invasion after over-expression of TIMPs using different gene de-livery vectors has been shown in vitro in gas-tric cancer cells and mammary carcinoma cells(156, 157). Overexpression in vitro of TIMP-2,which was delivered by a recombinant adeno-virus (AdTIMP-2), inhibited the invasion ofboth tumor and endothelial cells in three mu-rine models without affecting cell prolifera-tion (158). Its in vivo efficiency has been eval-uated in the LLC murine lung cancer model,the colon cancer C51 model, as well as in MDA-MB231 human breast cancer in athymic mice.Preinfection of tumor cells by AdTIMP-2 re-sulted in an inhibition of tumor establishmentin more than 50% of mice in LLC and C51models and in 100% of mice in the MDA-MB231 model. A single local injection ofAdTIMP-2 into preestablished tumors ofthese three tumor types reduced tumorgrowth rates by 60–80%, and the tumor-asso-ciated angiogenesis index by 25–75%. Lungmetastasis of LLC tumors was inhibited by�90%. In addition, AdTIMP-2-treated miceshowed a significantly prolonged survival inall the cancer models tested. These data dem-onstrate the potential of adenovirus-mediatedTIMP-2 therapy in cancer treatment.

5 Interventions 45

Page 46: Molecular Biology of Cancer - UC · PDF fileDepartments of Radiation Oncology/Cancer Biology Section Molecular and Cellular Biology ... 2 Molecular Biology of Cancer. are genetic mutations

6 ACKNOWLEDGMENTS

We would like to acknowledge the expert anddedicated assistance of Kim Nicolini withoutwhom this chapter would never have beencompleted. Her perseverance and insistencethat we keep pushing forward to the end aremuch appreciated.

REFERENCES1. M. H. Myers and L. A. Ries, Can. Cancer

J. Clin., 39, 21–32 (1989).2. R. Peto, IARC Sci. Publ., 39, 27–28 (1982).3. P. C. Nowell, Science, 194, 23–28 (1976).4. D. Stehelin, H. E. Varmus, J. M. Bishop, and

P. K. Vogt, Nature, 260, 170–173 (1976).5. H. Land, L. F. Parada, and R. A. Weinberg,

Nature, 304, 596–602 (1983).6. E. R. Fearon and B. Vogelstein, Cell, 61, 759–

767 (1990).7. H. Hennings, A. B. Glick, D. A. Greenhalgh,

D. L. Morgan, J. E. Strickland, T. Tennen-baum, and S. H. Yuspa, Proc. Soc. Exp. Biol.Med., 202, 1–8 (1993).

8. M. A. Nelson, B. W. Futscher, T. Kinsella, J.Wymer, and G. T. Bowden, Proc. Natl. Acad.Sci. USA, 89, 6398–6402 (1992).

9. T. Minamoto, M. Mai, and Z. Ronai, Carcino-genesis, 20, 519–527 (1999).

10. I. Tannock and R. P. Hill, The Basic Science ofOncology, Pergamon Press, New York, 1987.

11. J. S. Bertram, Mol. Aspects Med., 21, 167–223(2000).

12. E. C. Friedberg, G. C. Walker, and W. Siede,DNA Repair and Mutagenesis, American Soci-ety for Microbiology Press, Washington, DC,1995.

13. M. L. Slattery, W. Samowitz, L. Ballard, D.Schaffer, M. Leppert, and J. D. Potter, CancerRes., 61, 1000–1004 (2001).

14. M. Iwamoto, D. J. Ahnen, W. A. Franklin, andT. H. Maltzman, Carcinogenesis, 21,1935–1940 (2000).

15. Y. Guo, R. B. Harris, D. Rosson, D. Boorman,and T. G. O’Brien, Cancer Res., 60, 6314–6317(2000).

16. T. Bestor, A. Laudano, R. Mattaliano, and V.Ingram, J. Mol. Biol., 203, 971–983 (1988).

17. A. P. Bird and A. P. Wolffe, Cell, 99, 451–454(1999).

18. W. Doerfler, Annu. Rev. Biochem., 52, 93–124(1983).

19. T. Goto and M. Monk, Microbiol. Mol. Biol.Rev., 62, 362–378 (1998).

20. D. P. Barlow, Science, 270, 1610–1613 (1995).21. S. B. Baylin, M. Esteller, M. R. Rountree, K. E.

Bachman, K. Schuebel, and J. G. Herman,Hum. Mol. Genet., 10, 687–692 (2001).

22. R. Z. Chen, U. Pettersson, C. Beard, L. Jack-son-Grusby, and R. Jaenisch, Nature, 395,89–93 (1998).

23. M. O. Hiltunen, L. Alhonen, J. Koistinaho, S.Myohanen, M. Paakkonen, S. Marin, V. M.Kosma, and J. Janne, Int. J. Cancer, 70, 644–648 (1997).

24. M. F. Kane, M. Loda, G. M. Gaida, J. Lipman,R. Mishra, H. Goldman, J. M. Jessup, and R.Kolodner, Cancer Res., 57, 808–811 (1997).

25. S. J. Nass, J. G. Herman, E. Gabrielson, P. W.Iversen, F. F. Parl, N. E. Davidson, and J. R.Graff, Cancer Res., 60, 4346–4348 (2000).

26. C. M. Bender, J. M. Zingg, and P. A. Jones,Pharm. Res., 15, 175–187 (1998).

27. L. E. Lantry, Z. Zhang, K. A. Crist, Y. Wang,G. J. Kelloff, R. A. Lubet, and M. You, Carcino-genesis, 20, 343–346 (1999).

28. R. W. Ruddon,. Cancer Biology, 3rd ed., OxfordUniversity Press, New York, (1995).

29. J. C. Houck, V. K. Sharma, and L. Hayflick,Proc. Soc. Exp. Biol. Med., 137, 331–333(1971).

30. D. J. Bearss, L. H. Hurley, and D. D. Von Hoff,Oncogene, 19, 6632–6641 (2000).

31. F. Ciardiello, Drugs, 60, 25–32 (2000).32. I. Stamenkovic, Sem. Cancer Biol., 10, 415–

433 (2000).33. H. Nagase, Biol. Chem., 378, 151–160 (1997).34. A. Lochter and M. J. Bissell, Apmis., 107, 128–

136 (1999).35. B. Davidson, I. Goldberg, P. Liokumovich, J.

Kopolovic, W. H. Gotlieb, L. Lerner-Geva, I.Reder, G. Ben-Baruch, and R. Reich, Int. J.Gynecol. Pathol., 17, 295–301 (1998).

36. J. D. Knox, C. Wolf, K. McDaniel, V. Clark, M.Loriot, G. T. Bowden, and R. B. Nagle, Mol.Carcinog., 15, 57–63 (1996).

37. R. G. Chirivi, A. Garofalo, M. J. Crimmin, L. J.Bawden, A. Stoppacciaro, P. D. Brown, and R.Giavazzi, Int. J. Cancer, 58, 460–464 (1994).

38. A. E. Aplin, A. Howe, S. K. Alahari, and R. L.Juliano, Pharmacol Rev., 50, 197–263 (1998).

39. Z. Dong, R. Kumar, X. Yang, and I. J. Fidler,Cell, 88, 801–810 (1997).

40. J. P. Witty, S. McDonnell, K. J. Newell, P. Can-non, M. Navre, R. J. Tressler, and L. M. Matri-sian, Cancer Res., 54, 4805–4812 (1994).

Molecular Biology of Cancer46

Page 47: Molecular Biology of Cancer - UC · PDF fileDepartments of Radiation Oncology/Cancer Biology Section Molecular and Cellular Biology ... 2 Molecular Biology of Cancer. are genetic mutations

41. A. B. Pardee, Proc. Natl. Acad. Sci. USA, 71,1286–1290 (1974).

42. C. Hutchison and D. M. Glover, Cell Cycle Con-trol, IRL Press, Oxford, 1995.

43. B. T. Zafonte, J. Hulit, D. F. Amanatullah, C.Albanese, C. Wang, E. Rosen, A. Reutens, J. A.Sparano, M. P. Lisanti, and R. G. Pestell, FrontBiosci., 5, D938–D961 (2000).

44. J. W. Harper, Cancer Surv., 29, 91–107 (1997).45. W. S. el-Deiry, J. W. Harper, P. M. O’Connor,

V. E. Velculescu, C. E. Canman, J. Jackman,J. A. Pietenpol, M. Burrell, D. E. Hill, and Y.Wang, Cancer Res., 54, 1169–1174 (1994).

46. L. Liu, N. J. Lassam, J. M. Slingerland, D.Bailey, D. Cole, R. Jenkins, and D. Hogg, On-cogene, 11, 405–412 (1995).

47. T. Weinert, Cancer Surv., 29, 109–132 (1997).48. S. W. Lowe, H. E. Ruley, T. Jacks, and D. E.

Housman, Cell, 74, 957–967 (1993).49. S. N. Farrow, Biochem. Soc. Trans., 27, 812–

814 (1999).50. J. Wang and M. J. Lenardo, J. Cell Sci., 113,

753–757 (2000).51. M. Loeffler and G. Kroemer, Exp. Cell Res.,

256, 19–26 (2000).52. A. Gross, J. M. McDonnell, and S. J. Kors-

meyer, Genes Dev., 13, 1899–1911 (1999).53. A. Ashkenazi and V. M. Dixit, Curr. Opin. Cell

Biol., 11, 255–260 (1999).54. R. C. Seeger, G. M. Brodeur, H. Sather, A. Dal-

ton, S. E. Siegel, K. Y. Wong, and D. Ham-mond, N. Engl. J. Med., 313, 1111–1116(1985).

55. E. C. Friedberg, Cancer J. Sci. Am., 5, 257–263(1999).

56. G. L. Semenza, J. Appl. Physiol., 88, 1474–1480 (2000).

57. J. R. Gnarra, S. Zhou, M. J. Merrill, J. R. Wag-ner, A. Krumm, E. Papavassiliou, E. H. Old-field, R. D. Klausner, and W. M. Linehan, Proc.Natl. Acad. Sci. USA, 93, 10589–10594 (1996).

58. I. J. Fidler, and L. M. Ellis, Cell, 79, 185–188(1994).

59. D. R. Senger, S. J. Galli, A. M. Dvorak, C. A.Perruzzi, V. S. Harvey, and H. F. Dvorak, Sci-ence, 219, 983–985 (1983).

60. N. Ferrara, and W. J. Henzel, Biochem. Bio-phys. Res. Commun., 161, 851–858 (1989).

61. B. Millauer, M. P. Longhi, K. H. Plate, L. K.Shawver, W. Risau, A. Ullrich, and L. M.Strawn, Cancer Res., 56, 1615–1620 (1996).

62. M. Asano, A. Yukita, T. Matsumoto, S. Kondo,and H. Suzuki, Cancer Res., 55, 5296–5301(1995).

63. B. Olofsson, K. Pajusola, A. Kaipainen, G. vonEuler, V. Joukov, O. Saksela, A. Orpana, R. F.Pettersson, K. Alitalo and U. Eriksson, ProcNatl Acad Sci U S A, 93, 2576–81 (1996).

64. M. Orlandini, L. Marconcini, R. Ferruzzi andS. Oliviero., Proc. Natl. Acad. Sci. U S A, 93,11675–80 (1996).

65. M. G. Achen, M. Jeltsch, E. Kukk, T. Makinen,A. Vitali, A. F. Wilks, K. Alitalo and S. A.Stacker, Proc. Natl. Acad. Sci. U S A, 95,548–53 (1998).

66. E. Kukk, A. Lymboussaki, S. Taira, A.Kaipainen, M. Jeltsch, V. Joukov and K. Ali-talo, Development, 122, 3829–37 (1996).

67. S. Tessler, P. Rockwell, D. Hicklin, T. Cohen,B. Z. Levi, L. Witte, I. R. Lemischka, and G.Neufeld, J. Biol. Chem., 269, 12456–12461(1994).

68. W. Risau, Prog. Growth Factor Res., 2, 71–79(1990).

69. K. J. Kim, B. Li, J. Winer, M. Armanini, N.Gillett, H. S. Phillips, and N. Ferrara, Nature,362, 841–844 (1993).

70. N. Ueki, M. Nakazato, T. Ohkawa, T. Ikeda, Y.Amuro, T. Hada, and K. Higashino, Biochim.Biophys. Acta., 1137, 189–196 (1992).

71. N. Weidner, J. P. Semple, W. R. Welch, and J.Folkman, N. Engl. J. Med., 324, 1–8 (1991).

72. C. I. Bargmann, M. C. Hung, and R. A. Wein-berg, Cell, 45, 649–657 (1986).

73. M. Crul, G. J. de Klerk, J. H. Beijnen, and J. H.Schellens, Anticancer Drugs, 12, 163–184(2001).

74. A. B. Vojtek and C. J. Der, J. Biol. Chem., 273,19925–19928 (1998).

75. H. R. Bourne, D. A. Sanders, and F. McCor-mick, Nature, 349, 117–127 (1991).

76. M. R. Wallace, D. A. Marchuk, L. B. Andersen,R. Letcher, H. M. Odeh, A. M. Saulino, J. W.Fountain, A. Brereton, J. Nicholson, and A. L.Mitchell, Science, 249, 181–186 (1990).

77. D. Stokoe, S. G. Macdonald, K. Cadwallader,M. Symons, and J. F. Hancock, Science, 264,1463–1467 (1994).

78. P. Rodriguez-Viciana, P. H. Warne, R. Dhand,B. Vanhaesebroeck, I. Gout, M. J. Fry, M. D.Waterfield, and J. Downward, Nature, 370,527–532 (1994).

79. T. Sasaki, J. Irie-Sasaki, Y. Horie, K. Bach-maier, J. E. Fata, M. Li, A. Suzuki, D. Bou-chard, A. Ho, M. Redston, S. Gallinger, R.Khokha, T. W. Mak, P. T. Hawkins, L. Ste-phens, S. W. Scherer, M. Tsao, and J. M. Pen-ninger, Nature, 406, 897–902 (2000).

References 47

Page 48: Molecular Biology of Cancer - UC · PDF fileDepartments of Radiation Oncology/Cancer Biology Section Molecular and Cellular Biology ... 2 Molecular Biology of Cancer. are genetic mutations

80. M. Vidal, V. Gigoux, and C. Garbay, Crit. Rev.Oncol. Hematol., 40, 175–186 (2001).

81. G. Q. Daley and Y. Ben-Neriah, Adv. CancerRes., 57, 151–184 (1991).

82. D. Diekmann, S. Brill, M. D. Garrett, N. Totty,J. Hsuan, C. Monfries, C. Hall, L. Lim, and A.Hall, Nature, 351, 400–402 (1991).

83. M. J. Mauro, M. O’Dwyer, M. C. Heinrich, andB. J. Druker, J. Clin. Oncol., 20, 325–334(2002).

84. T. Hai, and T. Curran, Proc. Natl. Acad. Sci.USA, 88, 3720–3724 (1991).

85. M. Karin, Z. Liu, and E. Zandi, Curr. Opin.Cell. Biol., 9, 240–246 (1997).

86. K. F. Mitchell, J. Battey, G. F. Hollis, C. Moul-ding, R. Taub, and P. Leder, J. Cell. Physiol.,Suppl 3, 171–177 (1984).

87. P. J. Koskinen and K. Alitalo, Sem. CancerBiol., 4, 3–12 (1993).

88. T. C. He, A. B. Sparks, C. Rago, H. Hermeking,L. Zawel, L. T. da Costa, P. J. Morin, B. Vo-gelstein, and K. W. Kinzler, Science, 281,1509–1512 (1998).

89. L. Zheng and W. H. Lee, Exp. Cell Res., 264,2–18 (2001).

90. J. R. Bischoff, D. H. Kirn, A. Williams, C.Heise, S. Horn, M. Muna, L. Ng, J. A. Nye, A.Sampson-Johannes, A. Fattaey, and F. McCor-mick, Science, 274, 373–376 (1996).

91. S. Ikeda, S. Kishida, H. Yamamoto, H. Murai,S. Koyama, and A. Kikuchi, EMBO J., 17,1371–1384 (1998).

92. L. Simpson and R. Parsons, Exp. Cell Res.,264, 29–41 (2001).

93. M. Tamura, J. Gu, K. Matsumoto, S. Aota, R.Parsons, and K. M. Yamada, Science, 280,1614–1617 (1998).

94. G. Torre-Amione, R. D. Beauchamp, H. Koep-pen, B. H. Park, H. Schreiber, H. L. Moses, andD. A. Rowley, Proc. Natl. Acad. Sci. USA, 87,1486–1490 (1990).

95. S. Markowitz, J. Wang, L. Myeroff, R. Parsons,L. Sun, J. Lutterbaugh, R. S. Fan, E.Zborowska, K. W. Kinzler, and B. Vogelstein,Science, 268, 1336–1338 (1995).

96. J. E. Cleaver, J. Dermatol. Sci., 23, 1–11(2000).

97. H. T. Lynch, T. C. Smyrk, P. Watson, S. J.Lanspa, J. F. Lynch, P. M. Lynch, R. J. Cava-lieri, and C. R. Boland, Gastroenterology, 104,1535–1549 (1993).

98. P. Peltomaki and H. F. Vasen, Gastroenterol-ogy, 113, 1146–1158 (1997).

99. P. Greenwald, Sci. Am., 275, 96–99 (1996).

100. H. S. Wasan, M. Novelli, J. Bee, and W. F. Bod-mer, Proc. Natl. Acad. Sci. USA, 94, 3308–3313 (1997).

101. D. S. Alberts, M. E. Martinez, D. J. Roe, J. M.Guillen-Rodriguez, J. R. Marshall, J. B. vanLeeuwen, M. E. Reid, C. Ritenbaugh, P. A. Var-gas, A. B. Bhattacharyya, D. L. Earnest, andR. E. Sampliner, N. Engl. J. Med., 342, 1156–1162 (2000).

102. S. A. Leadon, Sem. Radiat. Oncol., 6, 295–305(1996).

103. M. Abdulla, and P. Gruber, Biofactors, 12,45–51 (2000).

104. G. S. Omenn, G. E. Goodman, M. D. Thorn-quist, J. Balmes, M. R. Cullen, A. Glass, J. P.Keogh, F. L. Meyskens Jr., B. Valanis, J. H.Williams Jr., S. Barnhart, M. G. Cherniack,C. A. Brodkin, and S. Hammar, J. Natl. CancerInst., 88, 1550–1559 (1996).

105. B. Reddy, A. Engle, S. Katsifis, B. Simi, H. P.Bartram, P. Perrino, and C. Mahan, CancerRes., 49, 4629–4635 (1989).

106. J. A. Baron, M. Beach, J. S. Mandel, R. U. vanStolk, R. W. Haile, R. S. Sandler, R. Rothstein,R. W. Summers, D. C. Snover, G. J. Beck, J. H.Bond, and E. R. Greenberg, N. Engl. J. Med.,340, 101–107 (1999).

107. E. Giovannucci, E. B. Rimm, A. Wolk, A. As-cherio, M. J. Stampfer, G. A. Colditz, and W. C.Willett, Cancer Res., 58, 442–447 (1998).

108. R. N. Dubois, S. B. Abramson, L. Crofford,R. A. Gupta, L. S. Simon, L. B. Van De Putte,and P. E. Lipsky, Faseb J., 12, 1063–1073(1998).

109. A. T. Koki, K. M. Leahy, and J. L. Masferrer,Expert Opin. Investig. Drugs, 8, 1623–1638(1999).

110. F. L. Meyskens Jr. and E. W. Gerner, Clin.Cancer Res., 5, 945–951 (1999).

111. D. A. Notterman, U. Alon, A. J. Sierk, and A. J.Levine, Cancer Res., 61, 3124–3130 (2001).

112. J. Yu, L. Zhang, P. M. Hwang, C. Rago, K. W.Kinzler, and B. Vogelstein, Proc. Natl. Acad.Sci. USA, 96, 14517–14522 (1999).

113. M. Hidalgo and S. G. Eckhardt, J. Natl. CancerInst., 93, 178–193 (2001).

114. P. D. Delmas, N. Engl. J. Med., 335, 1836–1837 (1996).

115. O. Teronen, P. Heikkila, Y. T. Konttinen, M.Laitinen, T. Salo, R. Hanemaaijer, A. Teronen,P. Maisi, and T. Sorsa, Ann. NY Acad. Sci.,878, 453–465 (1999).

Molecular Biology of Cancer48

Page 49: Molecular Biology of Cancer - UC · PDF fileDepartments of Radiation Oncology/Cancer Biology Section Molecular and Cellular Biology ... 2 Molecular Biology of Cancer. are genetic mutations

116. M. A. Schwalke, G. N. Tzanakakis, and M. P.Vezeridis, J. Surg. Res., 49, 164–167 (1990).

117. M. E. Barry, D. Pinto-Gonzalez, F. M. Orson,G. J. McKenzie, G. R. Petry, and M. A. Barry,Hum. Gene Therap., 10, 2461–2480 (1999).

118. M. A. Hickman, R. W. Malone, K. Lehmann-Bruinsma, T. R. Sih, D. Knoell, F. C. Szoka, R.Walzem, D. M. Carlson, and J. S. Powell, Hum.Gene Therap., 5, 1477–1483 (1994).

119. J. P. Yang, and L. Huang, Gene Therap., 3,542–548 (1996).

120. R. Niven, R. Pearlman, T. Wedeking, J. Mack-eigan, P. Noker, L. Simpson-Herren, and J. G.Smith, J. Pharm. Sci., 87, 1292–1299 (1998).

121. R. M. Bartholomew, A. F. Esser, and H. J. Mul-ler-Eberhard, J. Exp. Med., 147, 844–853(1978).

122. F. Bushman, Science, 267, 1443–1444 (1995).

123. S. V. Joang, E. B. Stephens, and O. Narayan inB. N. Fields, D. M. Knipe and P. M. Howley,Eds., Virology, Lippincott-Raven, Philadel-phia, PA, 1996, 1997–1996.

124. U. Blomer, L. Naldini, T. Kafri, D. Trono, I. M.Verma, and F. H. Gage, J. Virol., 71, 6641–6649 (1997).

125. J. C. Cusack Jr. and K. K. Tanabe, Surg. Oncol.Clin. North Am., 7, 421–469 (1998).

126. P. Seth, Gene Therapy for the Treatment ofCancer, Vol. 44–77, Cambridge UniversityPress, Cambridge, 1998.

127. V. N. Krasnykh, G. V. Mikheeva, J. T. Douglas,and D. T. Curiel, J. Virol., 70, 6839–6846(1996).

128. J. A. Roth and R. J. Cristiano, J. Natl. CancerInst., 89, 21–39 (1997).

129. A. Iida, S. T. Chen, T. Friedmann, and J. K.Yee, J. Virol., 70, 6054–6059 (1996).

130. E. W. Gerner, E. M. Hersh, M. Pennington,T. C. Tsang, D. Harris, F. Vasanwala, and J.Brailey, Int. J. Hyperthermia, 16, 171–181(2000).

131. T. Clackson, Curr. Opin. Chem. Biol., 1, 210–218 (1997).

132. P. L. Felgner and G. M. Ringold, Nature, 337,387–388 (1989).

133. N. S. Templeton, D. D. Lasic, P. M. Frederik,H. H. Strey, D. D. Roberts, and G. N. Pavlakis,Nature Biotechnol., 15, 647–652 (1997).

134. A. Kikuchi, Y. Aoki, S. Sugaya, T. Serikawa, K.Takakuwa, K. Tanaka, N. Suzuki, and H.

Kikuchi, Hum. Gene Therap., 10, 947–955(1999).

135. C. Meunier-Durmort, R. Picart, T. Ragot, M.Perricaudet, B. Hainque, and C. Forest, Bio-chim. Biophys. Acta, 1330, 8–16 (1997).

136. T. Tuting, W. J. Storkus, and M. T. Lotze, J.Mol. Med., 75, 478–491 (1997).

137. M. Lindauer, T. Stanislawski, A. Haussler, E.Antunes, A. Cellary, C. Huber, and M.Theobald, J. Mol. Med., 76, 32–47 (1998).

138. J. Banchereau, F. Briere, C. Caux, J. Davoust,S. Lebecque, Y. J. Liu, B. Pulendran, and K.Palucka, Annu. Rev. Immunol., 18, 767–811(2000).

139. B. M. Davis, O. N. Koc, K. Lee, and S. L. Ger-son, Curr. Opin. Oncol., 8, 499–508 (1996).

140. Z. Ram, Isr. Med. Assoc. J., 1, 188–193 (1999).

141. D. Klatzmann, P. Cherin, G. Bensimon, O.Boyer, A. Coutellier, F. Charlotte, C. Boccac-cio, J. L. Salzmann, and S. Herson, Hum. GeneTherap., 9, 2585–2594 (1998).

142. D. H. Sterman, K. Molnar-Kimber, T. Iyengar,M. Chang, M. Lanuti, K. M. Amin, B. K. Pierce,E. Kang, J. Treat, A. Recio, L. Litzky, J. M.Wilson, L. R. Kaiser, and S. M. Albelda, CancerGene Therap., 7, 1511–1518 (2000).

143. R. G. Crystal, E. Hirschowitz, M. Lieberman,J. Daly, E. Kazam, C. Henschke, D. Yankelev-itz, N. Kemeny, R. Silverstein, A. Ohwada, T.Russi, A. Mastrangeli, A. Sanders, J. Cooke,and B. G. Harvey, Hum. Gene Therap., 8, 985–1001 (1997).

144. C. Beltinger, W. Uckert, and K. M. Debatin, J.Mol. Med., 78, 598–612 (2001).

145. J. Nemunaitis, S. G. Swisher, T. Timmons, D.Connors, M. Mack, L. Doerksen, D. Weill, J.Wait, D. D. Lawrence, B. L. Kemp, F. Fossella,B. S. Glisson, W. K. Hong, F. R. Khuri, J. M.Kurie, J. J. Lee, J. S. Lee, D. M. Nguyen, J. C.Nesbitt, R. Perez-Soler, K. M. Pisters, J. B.Putnam, W. R. Richli, D. M. Shin, and G. L.Walsh, J. Clin. Oncol., 18, 609–622 (2000).

146. T. Waku, T. Fujiwara, J. Shao, T. Itoshima, T.Murakami, M. Kataoka, S. Gomi, J. A. Roth,and N. Tanaka, J. Immunol., 165, 5884–5890(2000).

147. D. L. Tait, P. S. Obermiller, A. R. Hatmaker, S.Redlin-Frazier, and J. T. Holt, Clin. CancerRes., 5, 1708–1714 (1999).

148. K. R. Birikh, P. A. Heaton, and F. Eckstein,Eur. J. Biochem., 245, 1–16 (1997).

149. Y. A. Zhang, J. Nemunaitis, and A. W. Tong,Mol. Biotechnol., 15, 39–49 (2000).

References 49

Page 50: Molecular Biology of Cancer - UC · PDF fileDepartments of Radiation Oncology/Cancer Biology Section Molecular and Cellular Biology ... 2 Molecular Biology of Cancer. are genetic mutations

150. H. J. Andreyev, P. J. Ross, D. Cunningham,and P. A. Clarke, Gut, 48, 230–237 (2001).

151. W. A. Marasco, Gene Therap., 4, 11–15 (1997).152. F. Griscelli, H. Li, A. Bennaceur-Griscelli, J.

Soria, P. Opolon, C. Soria, M. Perricaudet, P.Yeh, and H. Lu, Proc. Natl. Acad. Sci. USA, 95,6367–6372 (1998).

153. P. Blezinger, J. Wang, M. Gondo, A. Quezada,D. Mehrens, M. French, A. Singhal, S. Sulli-van, A. Rolland, R. Ralston, and W. Min, Na-ture Biotechnol., 17, 343–348 (1999).

154. D. Ma, R. D. Gerard, X. Y. Li, H. Alizadeh, andJ. Y. Niederkorn, Blood, 90, 2738–2746(1997).

155. H. L. Kong, D. Hecht, W. Song, I. Kovesdi,N. R. Hackett, A. Yayon, and R. G. Crystal,Hum. Gene Therap., 9, 823–833 (1998).

156. M. Watanabe, Y. Takahashi, T. Ohta, M. Mai,T. Sasaki, and M. Seiki, Cancer, 77, 1676–1680(1996).

157. D. F. Alonso, G. Skilton, M. S. De Lorenzo,A. M. Scursoni, H. Yoshiji, and D. E. Gomez,Oncol. Rep., 5, 1083–1087 (1998).

158. H. Li, F. Lindenmeyer, C. Grenet, P. Opolon,S. Menashi, C. Soria, P. Yeh, M. Perricaudet,and H. Lu, Hum. Gene Therap., 12, 515–526(2001).

Molecular Biology of Cancer50