23
[CANCER RESEARCH (SUPPL.) 51, 5023s-5044s, September 15. 1991] Chemical and Physical Carcinogenesis: Advances and Perspectives for the 1990s1 Curtis C. Harris2 Laboratory of Human Carcinogenesis, National Cancer Institute, N1H, Bethesda, Maryland 20892 Abstract Carcinogenesis is a multistage process driven by carcinogen-induced genetic and epigenetic damage in susceptible cells that gain a selective growth advantage and undergo clonal expansion as the result of activation of protooncogenes and/or inactivation of tumor suppressor genes. There fore, the mutational spectra of chemical and physical carcinogens in these critical genes are of interest to define endogenous and exogenous muta tional mechanisms. The p53 tumor suppressor gene is ideally suited for analysis of the mutational spectrum. Such an analysis has revealed evidence for both exogenous and endogenous molecular mechanisms of Carcinogenesis. For example, an informative p53 mutational spectrum of frequent G—»T transversions in codon 249 is found in hepatocellular carcinomas from either Qidong, People's Republic of China, or southern Africa. This observation links exposure to aflatoxin B,, a known cancer risk factor in these geographic regions, with a specific mutation in a cancer-related gene. Other studies indicate that abnormalities in genes controlling the cell cycle may cause genomic instability and increase the probability of neoplastic transformation. Finally, mechanistic understand ing of Carcinogenesis is leading to improved cancer risk assessment and to the identification of individuals at high cancer risk. Introduction Carcinogenesis is a mature multidisciplinary field of cancer research that has a rich history of scientific accomplishments achieved by epidemiológica! observations (1-3); development and exploitation of both animal models relevant to human cancer (4) and in vitro cell and tissue models including the use of human tissues (5, 6); and most recently, advances in molec ular genetics (7-II).3 Examples of conceptual advances and seminal observations in the areas of chemical and physical Carcinogenesis are listed in Table 1; advances in viral Carcino genesis are reviewed by Howley in this issue (12). Since the research accomplishments of Carcinogenesis have been extensively reviewed (13-25), this commentary will focus on selected topics that are especially relevant to current under standing of human Carcinogenesis and, in particular, to the molecular epidemiology of human cancer, an emerging field in cancer research. Multistage Carcinogenesis Carcinogenesis is a multistage process driven by carcinogen- induced genetic and epigenetic damage in susceptible cells that gain a selective growth advantage and undergo clonal expansion as the result of activation of protooncogenes and/or inactivation of tumor suppressor genes. The traditional view of carcinogen- esis (Fig. 1) is derived primarily from studies of animal models. The first stage of the carcinogenic process, tumor initiation, involves exposure of normal cells to chemical, physical, or microbial carcinogens that cause a genetic change(s) providing 1Presented at the Symposium "Discoveries and Opportunities in Cancer Research: A Celebration of the 50th Anniversary of the Journal Cancer Research." May 15, 1991, during the 82nd Annual Meeting of the American Association for Cancer Research, Houston, TX. 2To whom requests for reprints should be addressed, at Laboratory of Human Carcinogenesis, National Cancer Institute, N1H, Building 37, Room 2C05, Be thesda, MD 20892. 3 Due to space limitations and the breadth of this topic, comprehensive reviews will be frequently cited and are the source of additional citations. the initiated cells with both an altered responsiveness to their microenvironment and moreover exerts a selective clonal ex pansion advantage when compared to the surrounding normal cells (26-28). The initiated cells may have decreased respon siveness to the inter- and intracellular signals that maintain normal tissue architecture and regulate the homeostatic growth and maturation of cells. For example, initiated cells may be less responsive to negative growth factors, inducers of terminal cell differentiation and/or programmed cell death (4, 26, 29-35). Perturbation of the normal microenvironment by (a) physical means, e.g., wounding of the mouse skin or partial hepatectomy in rodents, (b) chemical agents, e.g., exposure of the mouse skin to certain phorbol esters or exposure of the rat liver to phénobarbital, (c) the inflammatory process, or (d) microbial agents, e.g., influenza virus enhancement of rodent lung carci- nogenesis or hepatitis viruses in human liver Carcinogenesis, may also foster such selective clonal expansion of the initiated cells (4, 17, 36, 37). Tumor promotion results in proliferation and/or survival of the initiated cells to a greater extent than normal cells and enhances the probability of additional genetic damage including endogenous mutations accumulating in the expanding popula tion of these cells. The probability of a subpopulation of initi ated cells converting to malignancy can be substantially in creased by their further exposure to DNA-damaging agents (38-40) that may activate protooncogenes (41) and/or inacti vate tumor suppressor genes. Malignant cells continue to ex hibit progressive phenotypic changes during tumor progression (42) and may exhibit intrinsic genomic instability that is man ifested by the abnormal number and structure of chromosomes, gene amplification, and altered gene expression (43-45). This classical view of two stage Carcinogenesis involving a mutation, tumor initiation, and an epigenetic change, tumor promotion, has been conceptually important but is also consid ered to be simplistic in that the number of independent genetic and epigenetic events may be 6 or more in certain types of cancer (46-48). Furthermore, chemical carcinogens may: (a) not be considered mutagens, termed nongenotoxic carcinogens (49); (b) have both genetic and epigenetic effects such as the ability to induce mutations and the property of decreasing DNA methylation, respectively (50) or increase cellular proliferation; and (c) have a linear or non-linear dose-response relationship with respect to tumor formation (51, 52). These and other observations have generated an active debate concerning both the contribution of endogenous mutagenic mechanisms, e.g., oxy-radical DNA damage, DNA depurination, DNA polymer- ase infidelity, and deamination of 5-methylcytosine, versus ex ogenous environmental mutagens and the value of animal bioas- says to identify carcinogens and to aid in human cancer risk assessment (53-63). This debate is not merely an academic one in that societal and regulatory decisions critical to public health are at issue. Carcinogen Metabolism and DNA Damage Many chemical carcinogens require metabolic activation gen erally to high energy electrophiles to exert their carcinogenic 5023s on April 2, 2020. © 1991 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Chemical and Physical Carcinogenesis: Advances …...CARCINOGENESIS Table 1 Examples of advances in chemical and physical carcinogenesis" A. Cancer etiology I. Life-style factors (tobacco

  • Upload
    others

  • View
    3

  • Download
    1

Embed Size (px)

Citation preview

Page 1: Chemical and Physical Carcinogenesis: Advances …...CARCINOGENESIS Table 1 Examples of advances in chemical and physical carcinogenesis" A. Cancer etiology I. Life-style factors (tobacco

[CANCER RESEARCH (SUPPL.) 51, 5023s-5044s, September 15. 1991]

Chemical and Physical Carcinogenesis: Advances and Perspectives for the 1990s1

Curtis C. Harris2

Laboratory of Human Carcinogenesis, National Cancer Institute, N1H, Bethesda, Maryland 20892

Abstract

Carcinogenesis is a multistage process driven by carcinogen-induced

genetic and epigenetic damage in susceptible cells that gain a selectivegrowth advantage and undergo clonal expansion as the result of activationof protooncogenes and/or inactivation of tumor suppressor genes. Therefore, the mutational spectra of chemical and physical carcinogens in thesecritical genes are of interest to define endogenous and exogenous mutational mechanisms. The p53 tumor suppressor gene is ideally suited foranalysis of the mutational spectrum. Such an analysis has revealedevidence for both exogenous and endogenous molecular mechanisms ofCarcinogenesis. For example, an informative p53 mutational spectrum offrequent G—»Ttransversions in codon 249 is found in hepatocellularcarcinomas from either Qidong, People's Republic of China, or southern

Africa. This observation links exposure to aflatoxin B,, a known cancerrisk factor in these geographic regions, with a specific mutation in acancer-related gene. Other studies indicate that abnormalities in genes

controlling the cell cycle may cause genomic instability and increase theprobability of neoplastic transformation. Finally, mechanistic understanding of Carcinogenesis is leading to improved cancer risk assessment andto the identification of individuals at high cancer risk.

Introduction

Carcinogenesis is a mature multidisciplinary field of cancerresearch that has a rich history of scientific accomplishmentsachieved by epidemiológica! observations (1-3); developmentand exploitation of both animal models relevant to humancancer (4) and in vitro cell and tissue models including the useof human tissues (5, 6); and most recently, advances in molecular genetics (7-II).3 Examples of conceptual advances and

seminal observations in the areas of chemical and physicalCarcinogenesis are listed in Table 1; advances in viral Carcinogenesis are reviewed by Howley in this issue (12).

Since the research accomplishments of Carcinogenesis havebeen extensively reviewed (13-25), this commentary will focuson selected topics that are especially relevant to current understanding of human Carcinogenesis and, in particular, to themolecular epidemiology of human cancer, an emerging field incancer research.

Multistage Carcinogenesis

Carcinogenesis is a multistage process driven by carcinogen-induced genetic and epigenetic damage in susceptible cells thatgain a selective growth advantage and undergo clonal expansionas the result of activation of protooncogenes and/or inactivationof tumor suppressor genes. The traditional view of carcinogen-esis (Fig. 1) is derived primarily from studies of animal models.The first stage of the carcinogenic process, tumor initiation,involves exposure of normal cells to chemical, physical, ormicrobial carcinogens that cause a genetic change(s) providing

1Presented at the Symposium "Discoveries and Opportunities in CancerResearch: A Celebration of the 50th Anniversary of the Journal Cancer Research."

May 15, 1991, during the 82nd Annual Meeting of the American Association forCancer Research, Houston, TX.

2To whom requests for reprints should be addressed, at Laboratory of Human

Carcinogenesis, National Cancer Institute, N1H, Building 37, Room 2C05, Bethesda, MD 20892.

3 Due to space limitations and the breadth of this topic, comprehensive reviewswill be frequently cited and are the source of additional citations.

the initiated cells with both an altered responsiveness to theirmicroenvironment and moreover exerts a selective clonal expansion advantage when compared to the surrounding normalcells (26-28). The initiated cells may have decreased responsiveness to the inter- and intracellular signals that maintainnormal tissue architecture and regulate the homeostatic growthand maturation of cells. For example, initiated cells may be lessresponsive to negative growth factors, inducers of terminal celldifferentiation and/or programmed cell death (4, 26, 29-35).Perturbation of the normal microenvironment by (a) physicalmeans, e.g., wounding of the mouse skin or partial hepatectomyin rodents, (b) chemical agents, e.g., exposure of the mouseskin to certain phorbol esters or exposure of the rat liver tophénobarbital, (c) the inflammatory process, or (d) microbialagents, e.g., influenza virus enhancement of rodent lung carci-nogenesis or hepatitis viruses in human liver Carcinogenesis,may also foster such selective clonal expansion of the initiatedcells (4, 17, 36, 37).

Tumor promotion results in proliferation and/or survival ofthe initiated cells to a greater extent than normal cells andenhances the probability of additional genetic damage includingendogenous mutations accumulating in the expanding population of these cells. The probability of a subpopulation of initiated cells converting to malignancy can be substantially increased by their further exposure to DNA-damaging agents(38-40) that may activate protooncogenes (41) and/or inactivate tumor suppressor genes. Malignant cells continue to exhibit progressive phenotypic changes during tumor progression(42) and may exhibit intrinsic genomic instability that is manifested by the abnormal number and structure of chromosomes,gene amplification, and altered gene expression (43-45).

This classical view of two stage Carcinogenesis involving amutation, tumor initiation, and an epigenetic change, tumorpromotion, has been conceptually important but is also considered to be simplistic in that the number of independent geneticand epigenetic events may be 6 or more in certain types ofcancer (46-48). Furthermore, chemical carcinogens may: (a)not be considered mutagens, termed nongenotoxic carcinogens(49); (b) have both genetic and epigenetic effects such as theability to induce mutations and the property of decreasing DNAmethylation, respectively (50) or increase cellular proliferation;and (c) have a linear or non-linear dose-response relationshipwith respect to tumor formation (51, 52). These and otherobservations have generated an active debate concerning boththe contribution of endogenous mutagenic mechanisms, e.g.,oxy-radical DNA damage, DNA depurination, DNA polymer-ase infidelity, and deamination of 5-methylcytosine, versus exogenous environmental mutagens and the value of animal bioas-says to identify carcinogens and to aid in human cancer riskassessment (53-63). This debate is not merely an academic onein that societal and regulatory decisions critical to public healthare at issue.

Carcinogen Metabolism and DNA Damage

Many chemical carcinogens require metabolic activation generally to high energy electrophiles to exert their carcinogenic

5023s

on April 2, 2020. © 1991 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Page 2: Chemical and Physical Carcinogenesis: Advances …...CARCINOGENESIS Table 1 Examples of advances in chemical and physical carcinogenesis" A. Cancer etiology I. Life-style factors (tobacco

CARCINOGENESIS

Table 1 Examples of advances in chemical and physical carcinogenesis"

A. Cancer etiologyI. Life-style factors (tobacco snuff or smoke) can cause human cancer.

2. Occupational agents and factors can cause human cancer.a. Nun (childlessness)b. Coal sootc. Sunlightd. Aromatic aminese. Benzenef. Ionizing irradiation

g. Asbestos

3. Mixtures of chemicals (coal tar) can cause cancer in experimental animals.

4. Specific chemicals, e.g., dibenzanthracene or 2-naphthylamine, are carcinogens in experimental animals.

5. Solid "inert" substances such as plastic can cause cancer when implanted s.c. in experimen

tal animals.

6. Naturally occurring compounds isolated from plants, e.g., croton oil, or marine organisms,e.g.. teleocidin B and okadaic acid, have cocarcinogenic and/or tumor-promoting activities inexperimental animals.

7. Naturally occurring chemicals such as alkaloids from plants, ¡-.K--bracken fern (Pleritliumaquilinum). cycad. and Senecio, and fungal toxins, e.g.. aflatoxin B,. can be carcinogens inexperimental animals.

8. yV-Nitrosamines can be carcinogenic in laboratory animals.

9. Cooking of red meat, fish, or specific amino acids can generate polynuclear aromatic hydrocarbons and, in some cases, heterocyclic amines that are carcinogenic in animal models.

10. Tobacco smoking and alcoholic beverages are human cancer risk factors and have multiplicative effects.

11. Exposure of the human fetus to ionizing radiation increases risk of leukemia.

12. Tobacco smoking and occupational exposure to asbestos increases risk of human broncho-genie carcinoma.

13. Dietary exposure to aflatoxin B, correlates with prevalence of human hepatocellular carcinoma in Africa.

14. Animal bioassays can identify chemical carcinogens, e.g., chloromethyl ethers and vinylchloride, prior to epidemiological evidence.

15. Carcinogen-macromolecular adducts can be used as molecular dosimeters including exposureto life-style and occupational carcinogens.

16. Transplacental exposure to diethylstilbestrol increases risk of human vaginal carcinoma.

17. Fiber dimension and length is correlated with carcinogenicity in animal models

Hill. 1759(329)Soemmerring, 1795 (330)Doll and Hill. 1950(331)Wynder and Graham, 1950(332)

Ramazzini. 1713(333)Pott, 1775(334)Thiersch, 1875(335)Rehn. 1895(337)DeLore and Borgman, 1928 (340)Harting and Hesse, 1879 (336)Trieben, 1902 (338)Clune!, 1910(339)Martland and Humphries, 1929 (341)Ministry of Labour and National Serv

ice, 1949(342)Wagner et al., 1960(343)

Yamagiwa and Ichikawa. 1918 (344)

Kennaway and Hieger, 1930 (345)Hueper et al.. 1938(346)

Turner. 1941 (347)

Berenblum, 1941 (348)Fujiki et al.. 1979(349)

Cook el al., 1950(355)Schoentalf/a/.. 1954(356)Laquer, 1964(357)Evans and Mason, 1965 (358)Carnaghan, 1965(359)Wogan, 1966(360)Wogan and Newberne. 1967 (361)

Magee and Barnes. 1956 (362)

Kuratsume, 1956(363)Lijinsky and Shubik, 1964 (364)Sugimura et al.. 1977(365)Kasai el al.. 1980(366)

Wynder el al.. 1957(367)Keller. 1967(368)Rothman and Keller, 1972 (369)

Stewart étal.,1958(370)

Selikofff/a/., 1968(371)

Alpert et al.. 1968(372)

Van Duuren el al.. 1968 (373)Viola el al., I97I (374)

Ehrenberg el al.. 1974(296)Tornqvist et al.. 1986 (298)Farmer elal.. 1986(300)

Herbst et al., 1977(375)

Stanton el al., 1981 (376)

B. Multistage carcinogenesis

1. Chemicals can cause cancer at sites distant from the point of application in experimentalanimals.

2. Metabolites of chemical carcinogens are predicted to be the active carcinogenic agent.

3. Carcinogenesis is a multifactorial process involving tumor initiation and promotion in animal models.

Sasaki and Yoshida. 1935 (377)

Boyland. 1935(378)

Rousand Kidd. 1941 (379)Berenblum. 1941 (348)Mottram. 1944(380)Berenblum and Shubik. 1949 (381)

5024s

on April 2, 2020. © 1991 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Page 3: Chemical and Physical Carcinogenesis: Advances …...CARCINOGENESIS Table 1 Examples of advances in chemical and physical carcinogenesis" A. Cancer etiology I. Life-style factors (tobacco

Table 1 Continued

4. Metabolic activation of some chemical carcinogens is essential for their carcinogenic activity.

5. Tumor promoters can inhibit cell-cell communication of mammalian cells in vitro.

6. A tumor promoter in experimental animals can specifically bind and activate a specificcellular receptor.

7. Oncogenes cooperate in neoplastic transformation of primary rodent cells.

8. Some tumor promoters act via the protein kinase C pathway for signal transduction.

9. Cooperation between oncogenes is demonstrated in transgenic animal models.

10. Overexpression of protein kinase C leads to morphological transformation of rodent cells.

11. Multiple genetic events in protooncogenes and tumor suppressor genes are associated withhuman carcinogenesis.

Millerand Miller. 1947(382)Miller et al., 1964(383)

Murray and Fitzgerald, 1979 (384)\ott\etal., 1979(385)

Driedgerand Blumberg, 1980 (386)Castagna et a!., 1982 (387)

Land et al., 1983(388)Ruley, 1983(389)

Davis and Czech. 1985 (390)Arroleo and Weinstein. 1985 (391)Ebeling et al., 1985(392)

Sinn étal.,1987(153)

Housey et al., 1988(393)Personsera/., 1988(394)

Volgelsteinera/., 1988(395)

C. Genetic and epigenetic mechanisms of carcinogenesis

1. Human cancer can be inherited.

2. Chromosomal abnormalities are predicted to be involved in cancer.

3. Carcinogenic X-rays are also mutagens.

4. Inbred mouse strains differ in their cancer incidence.

5. Cancer may arise by inactivation of both alíelesof a tumor suppressor gene.

6. Chemical carcinogens can be mutagens.

7. Chemical carcinogens may bind to DNA.

8. A specific chromosomal abnormality can be associated with human leukemia.

9. UV light causes a specific type of DNA damage, i.e., thymidine dimers (cyclobutane).

10. Carcinogenic potency of some chemical carcinogens correlates with their binding level toDNA in experimental animals.

11. Defective DNA repair may predispose to UV-induced human skin cancer.

12. Interindividual variation is observed in carcinogen metabolism and/or formation of carcino-gen-DNA adducts in human cells and tissues.

13. Normal cells are dominant in normal-tumor murine cell hybrids.

14. In vitro bacterial mutagenesis can be used to screen putative carcinogens.

15. RNA viral oncogenes have a normal cellular counterpart (protooncogene).

16. Data consistent with the autocrine growth factor theory are presented.

17. DNA from chemically transformed rodent cells contain an oncogene(s).

18. Specific chromosomal loss is associated with tumorigenic revenants of normal-carcinomahuman cell hybrids.

19. Chromosomal translocation can activate protooncogenes in human cells.

20. Oncogenes are cloned from human cancers.

21. raj oncogene contains a base substitution when compared to protooncogene.

Watkins, 1904(396)

Boveri, 1914(397)

Muller. 1927(398)

Forth et al., 1933(399)Strong, 1935(400)

Charles and Luce-Clausen, 1942 (401)Knudson, 1971 (95)Comings, 1973(402)Cavenee«a/.. 1983(96)

Auerbach et al., 1947 (403)Boyland and Horning, 1949 (404)

Boyland, 1952(405)Wheeler and Skipper. 1957 (406)Brookes and Lawley. 1960 (407)Farber and Magee. 1960 (408)

Nowell and Hungerford. 1960 (409)

Beukers and Berends, 1960 (410)

Brooks and Lawley, 1964 (411)

Cleaver, 1968(412)

Kuntzman et al., 1968 (413)Welch et al., 1968(414)Huberman and Sachs, 1973 (71)Harrisera/., 1976(415)

Harris et al.. 1969(416)

Õmese/a/., 1973(417)McCann ff a/., 1975(418)

Stehelin et al., 1976(419)

DeLarco and Todaro. 1978 (420)

Shihetal., 1979(421)

Stanbridge et al., 1981 (422)

Dalia-Pavera et al., 1982(423)deKlein et al., 1982(424)Shen-Ong et al.. 1982 (425)Taub« al.. 1982(426)

Goldfarb et al., 1982(427)Pulciani et al.. 1982(428)Shìhetal.. 1982(429)

Ready et al., 1982(430)Takinera/., 1982 (431)Taparowsky et al., 1982(432)

5025s

on April 2, 2020. © 1991 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Page 4: Chemical and Physical Carcinogenesis: Advances …...CARCINOGENESIS Table 1 Examples of advances in chemical and physical carcinogenesis" A. Cancer etiology I. Life-style factors (tobacco

CARCINOGENESIS

Table 1 Continued

22. Chemical carcinogens can activate ras protooncogenes in animal models.

23. Some mutant growth factor receptors are oncogenes in rodent cells.

24. Site-specific mutagenesis analysis of carcinogen-DNA adducts confirms predications of nu-cleotide misincorporation opposite specific adducts.

25. Transferred oncogenes can cause neoplastic transformation of human epithelial cells.

26. The human rctinoblastoma susceptibility (Kin gene is cloned.

27. Nuclear oncogenes can act as transcription factors.

28. Direct evidence of the tumor suppressor activity of the Rb gene in human cells.

29. pS3 is frequently mutated in diverse types of human cancers.

30. p53 functions as a tumor suppressor gene in rodent and human cells.

31. A candidate tumor suppressor gene on chromosome 18q may be involved in human coloncarcinogenesis

32. NF1. a GTPase-activating protein, may be a negative regulator of ras p2l and a tumorsuppressor gene

33. A candidate Wilm's tumor suppressor gene is cloned.

34. Germline />.\ìmutations are found in certain cancer-prone families.

35. Candidate tumor suppressor genes (MCC and APC) may be involved in familial adenoma-tous polyposis and colorectal carcinoma.

36. An environmental chemical carcinogen (aflatoxin B,) is linked with a specific mutation inthe p53 tumor suppressor gene in human hepatocellular carcinogenesis.

37. A candidate tumor suppressor gene (protein tyrosine phosphatase -y)on chromosome 3pmay be involved in human lung and renal carcinogenesis.

Balmain and Pragnell. 1983 (433)Sukumar et al., 1983 (434)

Downward et al., 1984 (435)

Loechler et al.. 1984(116)Preston et al.. 1986(436)

Yoakum et al., 1985(437)Kliim «-i«/..1985(438)

Friend et al., 1986(439)

Makiera/., 1987(440)Vogtefa/., 1987(441)Höhnumn et al., 1987 (442)

Huang étal..1988(147)

Baker et al., 1989(443)Nigroero/., 1989(127)Takahashi et al.. 1989(128)

Eliyahu et al., 1989(444)Finlay «a/.. 1989(445)Baker et al., 1989(443)

Fearonera/., 1990(446)

Cawthon et al., 1990 (447)Xuetal., 1990(448)Wallace étal..1990(449)

Callera/.. 1990(450)Gessler, 1990(451)

M:ilkiii <•/«/..1990(141)Srivastavaera/., 1990(142)Kinzler et al., 1991 (452)Kinzler et al., 1991 (453)Nishisho et al., 1991 (454)Graden et al., 1991 (455)

Hsufia/., 1991 (137)Bressac et al.. 1991 (138)

LaForgia et al., 1991 (456)

°These historical reviews have been useful references and are a source of additional examples of advances in carcinogenesis (350-354).

Initiation Promotion Conversion Progression

Fig. I. Carcinogenesis is a multistage process involving multiple genetic and epigeneticevents in protooncogenes. tumor suppressorgenes, and anti-metastasis genes.

•Defects in Terminal Differentiation

•Defects in Growth Control

•Resistance to Cytotoxicity

GeneticChange

inhiortion

NORMAL CELL

v —/ rïït$<'INITIATED

PRENEOPLASTICCELL LESIONMALIGNANT TUMORP*

'CLINICAL

CANCER•

Activation of Proto-Oncogenes

•Inactivation of Tumor Suppressor Genes

•Inactivation of Antimetastasis Genes

effects (64). Although interspecies differences have long beenknown in the metabolism of xenobiotics (65-69), the metabolicpathways of activation and the resultant carcinogen-DNA adducts are generally qualitatively similar among various animalspecies, including humans (6). These observations support the

qualitative extrapolation of carcinogenesis data from laboratoryanimals to the human situation. Quantitative and qualitativedifferences in cytochrome P-450 enzymes in various tissues (70)may be partly responsible for the divergent sites of cancerinduction among animal species; e.g., 4-aminobiphenyl causes

5026s

on April 2, 2020. © 1991 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Page 5: Chemical and Physical Carcinogenesis: Advances …...CARCINOGENESIS Table 1 Examples of advances in chemical and physical carcinogenesis" A. Cancer etiology I. Life-style factors (tobacco

HypothesisGeneration

ClinicalObservation

CARCINOGENESIS

In Vivo Studies In Vitro Studies

Fig. 2. Strategy for studies of molecularcarcinogenesis and molecular epidemiology toimprove interspecies extrapolation of carcinogenesis data and to identify high cancer riskindividuals. Epidemiology

Clinical Investigations

Molecular Epidemiology

Explant vCulture

CellCulture

(epithelial)

Molecules

Molecular Dosimetryof Carcinogen Exposure

Inherited Cancer Predisposition

MolecularCarcinogenesis

liver cancer in rodents and bladder cancer in dogs and humans(65) and may confound tissue site-specific extrapolation ofcarcinogenesis data among animal species. Quantitative differences in carcinogen metabolism and in the formation of carcin-ogen-DNA adducts are, however, found among different celltypes (71, 72), tissue types (6, 70, 73), and outbred individualsof the same species (74, 75). As discussed (see below) in thesection "Molecular Epidemiology of Human Cancer," these

quantitative differences may influence tissue site of cancer andan individual's cancer risk.

The influence of carcinogen metabolism on interspecies extrapolation of carcinogenesis data is becoming more well defined by the cloning of genes encoding enzymes involved inactivation and detoxication of chemical carcinogens. Investigators have recently cloned more than 150 CYP4 genes (76)

and other genes involved in carcinogen metabolism, such ashuman /V-acetyltransferase (77), human epoxide hydrolase (78,79), human NAD(P)H:menadione oxidoreductase (80), humanglutathione 5-transferases (81), and pig flavin-containing mon-ooxygenase (82). As discussed by Gonzalez et al. (70), interspecies differences in carcinogen metabolism may, in part, be dueto: (a) qualitative differences in CYP genes due to an absenceof a CYP expression in an animal species; (b) quantitative andqualitative variations in regulation of CYP genes among animalspecies; and (c) differences in CYP substrate or product specificity. Since examples of interspecies differences in each of the3 types noted above have been described, in vitro model systemsincluding those using human cells and tissues (6) or geneticallyengineered cells or rodents containing single or combinationsof human genes directing the expression of specific enzymesresponsible for carcinogen metabolism (70, 83) will serve as abridge between in vivo studies in animal models and the humansituation (Fig. 2).

The molecular and atomic analyses of the physical interactions between chemical carcinogens and DNA represent a majorachievement of cancer researchers in the last two decades (84).The information gleaned from these studies of carcinogen-DNA

4The abbreviations used are: CYP, cytochrome P-450; CHO, Chinese hamsterovary; BPDE, benzo(a)pyrene diol-epoxide; PKC, protein kinase C; TPA, 12-O-tetradecanoylphorbol-13-acetate; AHH, aryl hydrocarbon hydroxylase; GST, glutathione S-transferase; PAH, polycyclic aromatic hydrocarbon; HPLC, highperformance liquid chromatography; PTPase y, protein-tyrosine phosphatase y.

adducts has provided a mechanistic explanation of the muta-genicity of many carcinogens, including their mutational spectra, and a rationale for improved carcinogen exposure assessment in molecular epidemiological studies of human cancer.

Critical DNA Targets: Protooncogenes and TumorSuppressor Genes

Protooncogenes are normal cellular genes that, when inappropriately activated as oncogenes, cause dysregulation ofgrowth and differentiation pathways and enhance the probability of neoplastic transformation. Carcinogens can cause thegenetic changes that can lead to activation of protooncogenes,including base substitution mutations (84), chromosomal trans-locations (85), and gene amplification (45, 86). For example,ras protooncogenes are activated primarily at codons 12, 13,and 61 by base substitution mutations caused by chemical andphysical carcinogens in both cultured mammalian cells andanimal models (87-90) including transplacental exposure tochemical carcinogens (91). Mutation of the Ha-ras protooncogene is an early event in rodent models of skin and mammarycarcinogenesis (87, 92). In addition, the v-Ha-ras transgene cansubstitute for the initiation step in mouse skin carcinogenesisin transgenic mice (93, 94). This animal model should be usefulfor investigating the molecular mechanisms of tumor promotersand antipromoters.

In contrast to protooncogenes, tumor suppressor genes arenormal cellular genes that, when inappropriately inactivated,cause dysregulation of growth and differentiation pathways andenhance the probability of neoplastic transformation. Based on

Table 2 Examples of functions of putative tumor suppressor genes

Induce terminal differentiationMaintain genomic stabilityTrigger senescenceInduce programmed cell deathRegulate cell growth

Signal transducers of negative growth factorsRegulators, e.g., PTPase7. of tyrosine kinases

Inhibit proteasesAlter DNA methylase activityModulate histocompatibility antigensRegulate angiogenesisFacilitate cell-cell communication

5027s

on April 2, 2020. © 1991 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Page 6: Chemical and Physical Carcinogenesis: Advances …...CARCINOGENESIS Table 1 Examples of advances in chemical and physical carcinogenesis" A. Cancer etiology I. Life-style factors (tobacco

CARCINOGENESIS

the multifunctional classes of tumor suppressor genes (Table 2)their number may equal that of the oncogenes. Loss of functionof the products of both alíelesis the basis of the 2-hit hypothesisand the recessive nature of the genetic process (95). In additionto inactivation of both alíelesby mutational mechanisms (basesubstitution mutations, deletions, chromosomal nondisjunc-tion, and recombination) (96), inactivation of a normal alíeleby genomic imprinting (97) and its gene product by an increasedrate of proteolytic digestion (98) or dominant negative mechanisms (99) have been proposed. Based on target size theory, theinactivation of a single alíeleof a tumor suppressor gene shouldhave an intrinsically higher probability than activation of aprotooncogene, e.g., one of the ras protooncogenes in whichmutations in only a few specific codons will lead to activation.The requirement for inactivation of both alíelesof a tumorsuppressor gene counterbalances this probability except in thosecases in which an inactivated alíeleis inherited or the mechanism of inactivation is via a dominant negative mechanism, asproposed for the p53 tumor suppressor gene (99, 100).

Ionizing irradiation and carcinogenic hormones, metals, aldehydes, or fibers such as asbestos are generally inefficient inthe production of point mutations at the gene level but aremore efficient at causing chromosomal abnormalities and losses(62). Therefore, one would speculate that tumor suppressorgenes would be targeted by these carcinogens more readily thanprotooncogenes. Whereas many electrophilic metabolites ofchemical carcinogens form carcinogen-DNA adducts and causepoint mutations, chemical carcinogens also cause chromosomalabnormalities and micronuclei. Thus, both cytogenetic andmutagenesis assays have been developed to assess these mutational changes (44, 101-103).

Mutational Spectrum

Since mutations are largely responsible for activating protooncogenes and inactivating tumor suppressor genes, the mutational spectra of chemical and physical carcinogens are ofinterest to define endogenous and exogenous mutational mechanisms. Studies using prokaryotic and simple eukaryotic assaysand site-specific mutagenesis assays (104, 105) have shown thateach carcinogenic agent produces a "fingerprint" (106) linking

specific types and locations of DNA adducts with the mutational spectrum of the agent. Both exogenous genes, which areintroduced into the appropriate host cell by a shuttle vector(105, 107-110), and endogenous genes (106) such as adeninephosphoribosyltransferase (aprt), dihydrofolate reducíase(dhfr), and hypoxanthine-guanine phosphoribosyl transferase(hprt) have been utilized in the eukaryotic studies. These assayswill underestimate the mutational frequency due to deletions,chromosomal nondisjunction, and frame-shift mutations because other genes essential for cell survival may be adjacent tothe hemizygous nonessential detector gene. In addition, certainmutations may cause either clonal expansion or inhibition ofthe mutant cell and further complicate the interpretation of themutational spectrum.

In both exogenous and endogenous types of target genes, thecarcinogen-induced mutational spectra can be compared to thespontaneous mutational spectrum. The majority of spontaneous mutations analyzed in these assays are single base substitutions. However, interesting differences have been observedin the spontaneous mutational spectrum in the endogenous aprtof CHO cells compared to bacterial cells and exogenous genescarried by shuttle vectors in mammalian cells (111). Transitionsof G:C—»A:Tare frequent in CHO cells suggesting deaminationof 5-methylcytosine and/or DNA replication errors, whereasdeletions and insertions are more frequent in the other assaysystems. A comparison of the spontaneous mutational spectrumat the aprt locus in CHO cells with the spectra induced byionizing radiation, UV radiation or benzo(a)pyrene diol epox-ide is shown in Table 3. The mutational spectrum of UV lightis consistent with the mutagenesis models of the promutageniccyclobutane and pyrimidine:pyrimidone (6:4) photoproductsinduced by UV light. As noted previously, ionizing radiation ismost efficient in causing deletions. BPDE, which binds to the2-amino group of G, produces the predicted G:C—»T:Atrans-versions; a similar mutational spectrum for BPDE has beenobserved in human cell assays (112). Polycyclic aromatic hydrocarbons can also cause mutations at regions of the genesencoding pre-mRNA consensus splice sites, resulting in mRNAsplicing defects. For example, mutations affecting splicing inthe CHO dhfr gene were preferentially induced by 3«,4ß-dihy-droxy-1 a,2«-epoxy-1,2,3,4-tetrahydrobenzo(c)phenanthrene(113). The hprt locus is of particular interest because of thepotential to compare mutational spectra obtained from studiesusing cultured cells experimentally exposed to carcinogens withthose observed in human lymphocytes obtained from donorsexposed in vivo to environmental carcinogens (114).

The molecular analysis of mutationally activated ras protooncogenes in animal models of carcinogenesis generally supportsthe concept of mutational spectrum reflecting the DNA adduciformation and mutagenic activiiies elicited by specific chemicalcarcinogens. For example, the mulalions found in activated rasprotooncogenes associated with tumors of rodents exposed loIhe melhylaling AAnilroso compounds are predominancyG:C—»A:Tbase subslitutions (Table 4); these are likely due lomelhylalion of deoxyguanine al ihe Oh posilion followed bymispairing wilh ihymine during DNA synthesis (92, 110, 115-117). Although there are several deoxyguanine residues in rascodons lhal would generale a transforming protein if substi-luled wilh adenine, Ihe animal experimenls have revealed lhatthe mutalions occur overwhelmingly al only one of the "possible" mutation sites. However, preferential mutalion siles are

within the admittedly narrow range of possibilities for aclivalingras genes. Unexplained mutational specificities have been observed in other experimental systems (118-120) and may reflectthe differences in the spectrum of promulagenic carcinogen-DNA adducls, specificily of DNA repair enzymes (121), and/or Ihe resullanl amino acid changes are eilher silent or lethal.

Allhough the mutalional speclrum of ras prolooncogenes inrodent tumors caused by methylaling carcinogens is consistent

Table 3 Percentage distribution of mutations in the endogenous aprt locus, including multiple events in Chinese hamster ovary-cells"

TransitionsMutagenSpontaneous

UV radiationIonizing radiationBenzo(a)pyrene diol epoxideG:C-»A:T71615A:T-«:C0

260G:C—T:A13

S662TransversionsG:C—

C:G3

10614A:T—

C:G0

5190A:T-»T:A7

12139Deletions6

2315Insertions0

205

'Adapted from Ref. 106.

5028s

on April 2, 2020. © 1991 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Page 7: Chemical and Physical Carcinogenesis: Advances …...CARCINOGENESIS Table 1 Examples of advances in chemical and physical carcinogenesis" A. Cancer etiology I. Life-style factors (tobacco

CARCINOGENESIS

Table 4 Examples of predominant base substitution mutations in ras protooncogenes in rodent tumors

ConditionSpontaneousMethylating

carcinogen''Tetranitromethane

Benzo(a)pyreneSkin

(H-ras)!G35—

»A'(NMU,

MNNG)MouseLiver

Lung Mammary Skin(H-ras) (K-ras) (H-ras)(H-ras)C'»-.A

G3!-»TG3'^A/

*

(NNK, DMN)Liver

Lung(K-ras)(K-ras)G3'-^A*RatMammary(H-ras)G35—A*(NMU)Kidney(H-ras)(DMN-OMe)Esophagus(H-ras)G3'-»AJ(MB)

7,12-DimethylbenzanthraceneAflatoxin B,

Benzidine and congeners

Urethan Ai«_T» An

" Ref. 457.* Ref. 458.' Ref. 459.''NMU, jV-methyl-A"-nitrosourea; MNNG, /V-methyl-iV'-nitrosoguanidine; NNK, 4-(methylnitrosamine)-l-(3-pyridyl-l-butanone), DMN, A'-nitrosodimethyla-

mine; DMN-OMe, methyl(methoxymethyl)nitrosamine: and MB, jV-nitrosomethylbenzylamine.' Ref. 87./Ref. 460.'Ref. 461.* Ref. 117.1Ref. 462.' Ref. 467.* Ref. 463.'Ref. 150." Ref. 464."Ref. 139.°Ref. 465.' Ref. 466.* R. W. Wiseman, personal communication.'Ref. 139.

Table 5 Examples of human cancers with base substitution mutations in the pS3 tumor suppressor gene"

V\orldwidecancerburden(rank)*234781112CancerLungBreastColonEsophagusLiverBladderLeukemia

andlymphomasSarcomasBrainNo.

ofmutationsdetectedin tumor

celllinesandtumors^43"31393719'15531220Mutational

'hotspots"(codon)273175.248273,

282249273Mutations

atA:T65712131903G:C372632251712341217CpG-TpG74267052579G:C-A:T121331163730815G:C-T:A19709142221G:C-C:G661013221

°Hollstein et al. (132).* From Muir (468).' Only mutations in exons 5, 6, 7. and 8 are included in this analysis. Tumor cell line mutations are included in the analysis with the understanding that some of

these may have arisen in vitro.d Chiba et al. (136) reported a higher p53 mutation frequency in lung squamous cell carcinoma than in other non-small cell lung carcinomas.' Frequency and mutational spectrum may vary considerably with geographic origin of tumor.

with the formation of the promutagenic 06-melhyldeoxyguano-sine and mutational spectrum caused by A/-nitroso-/V'-methyl-

urea in the aprt gene of CHO cells (106), an alternative hypothesis is that the mutant ras genes preexist in certain cells andthat exposure to the methylating agent facilitates clonal expansion of these mutant cells. This latter hypothesis has been testedby investigating the induction of rodent tumors by chemicalcarcinogens that form DNA adducts with differing chemicalspecificity. As seen in Table 4, the mutational spectra werehighly correlated with each chemical carcinogen and reflectedthe predicted base substitution (84), i.e., G:C—»T:Atransversionfor benzo(a)pyrene which forms predominantly the /V2-BPDE-deoxyguanosine adduci (122) and A:T—>T:Atransversion for7,12-dimethylbenzanthracene which forms predominantly theA*-7,12-dimethyIbenzanthracene diol-epoxide-deoxyadenosine

adduci (123).

The mutalional speclra of aclivaled ras prolooncogenes inlumors and preneoplaslic lesions caused by chemical carcinogens in animal models (Table 4) may prove lo be inslructive inthe inlerprelalion of mulalions in prolooncogenes and tumorsuppressor genes found in human cancers (Table 5). The spectraof Ki-ras protooncogene mutations in human adenocarcinomasvary according lo lissue sile; e.g., G:C—»T:Atransversions occurin lung tumors, whereas G:C—»A:Ttransit ions predominale incolonie lumors. The inlerprelalion of Ihese results, however,may be confounded by sponlaneous mulalions engendered byeilher endogenous mulagens or DNA polymerase errors. Astwo examples, 8-hydroxyguanine, caused by oxy radical damage(124) and DNA depurination, can produce G:C—»T:Airansver-sions, and DNA polymerase infidelity will also produce G:C—»T:A transversions as well as olher changes (125).

The p53 lumor suppressor gene is ideally suiled for analysis5029s

on April 2, 2020. © 1991 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Page 8: Chemical and Physical Carcinogenesis: Advances …...CARCINOGENESIS Table 1 Examples of advances in chemical and physical carcinogenesis" A. Cancer etiology I. Life-style factors (tobacco

CARCINOGENESIS

of mutational spectrum: (a) p53 is well conserved in evolution(126) and 5 domains are >90% homologous in DNA sequenceamong humans and rodents that are frequently used in animalmodels of carcinogenesis; (b)p53 is mutated in diverse types ofhuman cancer (127-131) and, where examined, cancers inlaboratory animals5"7; (c) a wide spectrum of mutational types

and codon sites have been observed that presumably defineregions of the p53 protein likely to be essential for its biologicalactivity in cell cycle control, in tumor suppression, and for itsinteraction with other cellular and viral proteins. A recentreview (132) has concluded that the majority of the mutationsin human tumors occur in the evolutionarily highly conserveddomains in exons 5-8 of the p53 gene, and the missensemutations are predominantly transitions at G:C base pairs andare almost all (>95%) at amino acids that are entirely conservedin mouse, rat, monkey, and human. Mutational spectra alsovary among cancer types (Table 5). The G:C—»A:Ttransitionsare the most frequent in colon tumors and 68% of the mutationsoccur at CpG dinucleotides. These findings are consistent withthe endogenous mutational mechanism due to deamination of5-methylcytosine residues found at CpG dinucleotides in themammalian genome. More than one-half of the p53 mutationsin colon tumors are at "hot spots," codons 175, 248, 273, or

282, each of which is a CpG dinucleotide.In contrast to colon tumors, CpG dinucleotides are less

frequently sites of mutations in most other types of humancancers. Whereas G:C—>T:Amutations are rare in colon tumors, these transversions are significantly more common inbreast and lung cancers. The G:C—>T:Amutations in Ki-rojprotooncogene are also found in human lung adenocarcinomas( 133-135) and in rodent lung cancers caused by benzo(a)pyrene(Table 4). Interestingly, the p53 mutational spectra differ insmall cell lung cancer and non-small cell lung cancer; G:C—»

T:A transversions are common in the latter group of cancers(136). Whereas CpG—»TpGmutations occur with intermediatefrequency in esophageal cancers, 36% of the mutations are atA:T pairs which may be due in part to DNA depurination and/or exposure to chemical carcinogens such as urethan (Table 4),a contaminant of certain alcoholic beverages.

The most striking p53 mutational spectrum is found in hep-atocellular carcinomas from either Qidong, People's Republic

of China (137) or southern Africa (138). Eleven of 12 basesubstitution mutations in 26 tumors were at a single site, the3rd base position of codon 249, and all but 1 were G:C—>T:Atransversions. One additional mutation at codon 157 was alsoa G:C—>T:Atransversion. These tumors were from patientswho live in geographic areas where aflatoxin B, and hepatitisB virus are major risk factors. Aflatoxin B, is a naturallyoccurring hepatocarcinogen that forms /V7-deoxyguanosine pro-

mutagenic adducts and induces primarily G:C—»T:Atransversions and G:C—»A:Ttransitions in experimental systems (139)(Table 4). Analysis of liver tumors from geographical areaswhere aflatoxin BI is considered not to be a significant riskfactor will be instructive in determining whether exposure tothis fungal carcinogen may be responsible for these p53mutations.

An analysis of mutational spectra in human tumors frompopulations with differing risk factors may provide insight in amanner similar to those of the studies in experimental animalsdescribed above (Table 4). In addition to further studies of liver

'' A. Balmain, personal communication.' A. Klein-Szanto. personal communication.7G. Wogan, personal communication.

cancer, the mutational spectra of Ki-ras and/or the p53 genecould be compared to bronchogenic cancers, including the useof paraffin-embedded, fixed archival tissue, from cigarettesmokers versus nonsmokers, and from workers in high riskoccupations, e.g., uranium mining, mustard gas production,asbestos mining, and chloromethyl ether production.

Selective Clonal Expansion

The majority of human cancers and those induced by chemical and physical carcinogens in animal models are unicellularin origin (43, 140). The clonal nature of neoplasia is thecornerstone in the concept of multistage carcinogenesis andprovides the basis for current strategies designated to elucidatethe component genetic changes. If all of the clonally derivedcells in a tumor, although heterogeneous in other aspects (discussed below), have in common the critical genetic lesion(s)that triggers and/or drives their selective clonal growth thenthe identification of this lesion(s) is a realistic goal.

Recent data suggest that alterations in thep53 and Rb tumorsuppressor genes represent such critical genetic lesions. Forexample, germline mutations in the Rb gene predispose toretinoblastoma (95, 96) and similar mutations in the p53 geneconfer an inherited susceptibility to cancers of the breast, softtissues, brain, and other tissues (141, 142). In vitro transfectionof wild type p53 into neoplastic colon (143), bladder (144),brain (145), and bone ( 146) cell lines containing an endogenousmutant p53 gene suppresses cellular growth and/or tumori-genicity. Similar transfection studies using putative wild typeRb have also suppressed, to varying degrees, cell growth (147,148). Although the mechanism(s) of growth retardation of thesecell lines by p53 or Rb is uncertain, the inhibition of the cellcycle progression into S phase following induction of wild typep53 is accompanied by selective down-regulation of mRNA andprotein expression of proliferating cell nuclear antigen, a cofac-tor of DNA polymerase ¿,an important component of DNAreplication (149).

Studies of animal models suggest that certain activated pro-tooncogenes may also be candidate critical genes. Mutations inthe Ha-ras protooncogene are early genetic events in mouseskin carcinogenesis (150) and rat mammary carcinogenesis(151, 152). Certain oncogenes impart, as transgenes, an "initiated" state (93, 94), and cooperation among oncogenes has

been shown to increase cancer risk in transgenic mouse models(153). Whereas germline mutations occur in tumor suppressorgenes that increase cancer risk in the affected individuals (141,142), germline mutations that activate protooncogenes in humans have yet to be described. The success of transgenic mousestudies argues against the possibility that such mutations wouldlead to fetal death and thus not be observed in the humanpopulation.

Activated protooncogenes can modulate the responsivenessof mammalian cells to their microenvironment and their cellular neighbors. For example, rodent cells transformed by certaintransfected oncogenes have defective intercellular communication (154) and the EIA oncogene induces resistance to thegrowth-inhibitory effects of transforming growth factor ß,(155). When Ha-ras, Ki-ras, or the combination of c-myc andc-ra/are transferred into SV40 T "immortalized" human bron

chial epithelial cells (BEAS-2B), the cells had a reduced responsiveness to growth inhibition by either transforming growthfactor type ßor serum (156-158)* and thus have potentially

* Unpublished results.

5030s

on April 2, 2020. © 1991 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Page 9: Chemical and Physical Carcinogenesis: Advances …...CARCINOGENESIS Table 1 Examples of advances in chemical and physical carcinogenesis" A. Cancer etiology I. Life-style factors (tobacco

CARCINOGENESIS

gained a selective clonal expansion advantage. Antisense oli-gonucleotides or expression vectors producing antisense DNAfragments of the mRNAs of these oncogenes should be usefulin assessing the direct role of raí,myc, or ra/in the modulationof growth and differentiation in this in vitro model system ofhuman bronchial epithelial carcinogenesis.

The molecular mechanisms responsible for selective growthadvantage of initiated cells are likely to be numerous and involvecell-cell interactions (154), cell-matrix interactions (159), cell-

growth factor interactions (34, 160) and intrinsic aberrationsin regulation of the cell cycle (161), terminal differentiation(41), and programmed cell death (162). Deregulated expressionof Bcl-2, a novel inner mitochondrial membrane protein, extends the survival of lymphoid cells presumably by blockingprogrammed cell death, apoptosis, and, as a hybrid minigenewith an immunoglobulin gene, increases the probability oflymphoma in transgenic mice (163). Epstein-Barr viral lategene products may also extend the survival of B-lymphocytesby blocking their death by apoptosis (164). An example of astudy that has identified candidate mutant genes and/or reversed the neoplastic state by transfecting the wild type gene isthe transfection of «5and /3¡integrin complementary DNA intomalignant CHO cells with a much decreased intrinsic level ofthese cognate integrins leads to suppression of tumorigenicity(165). Indirect proof of mechanisms leading to abnormal cell-growth factor interactions include interruption of signal trans-duction pathways of positive growth factors by use of neutralizing antibodies to either growth factors, e.g., gastrin-releasingpeptide (166), or their cell membrane receptors, e.g., epidermalgrowth factor receptor (167), resulting in decreased tumorigenicity of the human carcinoma cells injected into athymicnude mice. Transfection of antisense oligonucleotides or vectorsexpressing antisense oligonucleotides of certain oncogenes maylead to a reversal of certain in vitro phenotypes, such as softagar growth or transformed morphology, and in some casescause a decrease in tumorigenicity (168-172).

Protein kinase and protein phosphatases are responsible forthe reversible phosphorylation of cellular proteins that controlgrowth and differentiation of mammalian cells (173,456). PKCis a multigene family with at least 8 members that have differential expression in specific tissue types and functions in signaltransduction (28, 174). Exposure of certain cells to the tumorpromoter TPA causes PKC to translocate to cellular membranes and the resultant activation of PKC isoforms is associated with rapid induction of expression of nuclear protoonco-genes c-jun and c-fos (175). Since exposure to TPA produces

many phenotypic responses that are cell type dependent (176),and activated PKC modulates cytosolic calcium concentrationand phosphorylates multiple protein substrates, the signaltransduction pathways involving PKC are difficult to delineate(28). One hypothesis of the mechanistic role of PKC in tumorpromotion in mouse skin carcinogenesis is that certain proteins,when phosphorylated at specific sites directly or indirectly byPKC, activate signal transduction pathway(s) positive for cellgrowth. Assuming that the phosphorylated protein(s) is in thehypothetical active state, dephosphorylation at specific siteswould lead to inactivation. Therefore, inhibitors of the responsible protein phosphatase(s) would increase the steady statelevels of the critical phosphoprotein(s) in the activated stateand would be candidate tumor promoters. Although the phos-phoprotein(s) critical in tumor promotion has not as yet beenidentified, this latter hypothesis is supported by the recentdiscovery of non-TPA-like tumor promoters (177) such as

okadaic acid, a potent inhibitor of protein phosphatases I andIIA (178, 179). The interpretation of the above studies isrestricted because of the intrinsic limitations of experimentaldesigns using agonists and/or inhibitors that frequently havemultiple nonspecific effects. Since members of the PKC andthe protein phosphatase multigene families are being cloned,more powerful genetic strategies are becoming available.

Genomic Instability

Normal human somatic cells maintain a diploid karyotypeand have a mutation frequency of about 10~8 for hemoglobinvariants (point mutations), about 10~7 for hprt variants (meas

ures many classes of mutations, but the assay does not detectmitotic recombination), and about 10~5 for glycophorin A or

HLA variants (measures most classes of mutations includingsomatic recombination, gene conversion, or chromosomal non-disjunction) (114, 180, 181).

The potential sources for these observed mutation frequenciesinclude exposure to "background" levels of environmental

chemical and physical agents and endogenous mechanisms (55)such as: (a) the chemical instability of DNA, e.g., depurination[one can calculate that each cell undergoes IO4 depurinations/day (125)] or deamination of 5-methylcytosine to yield thymi-dine (182, 183); (b) free radicals of oxygen, e.g., hydroxyl ions[from measurements of 8-hydroxyguanine and thymine glycolin urine, about IO4 oxy radical-induced DNA lesions occur in

each human cell per day (54)]; and (c) DNA replication errors.Imbalances in deoxynucleoside triphosphate pools (184) andmutations in DNA polymerase «(185) are examples of mechanisms leading to infidelity of DNA synthesis. Inactivation ofRAD18, a putative DNA repair gene in Saccharomyces cerevis-iae, leads to an increase in the frequency of G:C—>T:Atrans-versions ( 186). Considering the multiplicity of proteins involvedin DNA synthesis, DNA repair, mitosis, and the cell cycle, thenumber of potential gene targets in somatic cells is large andcould result in the mutator phenotype (43, 55, 187) (Table 6)that could increase the probability of both neoplastic transformation and the generation of more malignant subclones duringtumor progression (42, 188). If such mutations occur in germcells and are nonlethal, they could lead to congenital abnormalities and increased cancer risk in an inherited manner. Thehypothesis that genomic instability may have an inherited basisand be associated with increased cancer risk is supported bystudies of Bloom's syndrome, where an increase in cytogenetic

end points is observed (189), and of colonie carcinomas, wherea significant increase in allelic deletions are found in tumorsfrom patients who have first degree relatives with cancer (190).

Cytogenetic studies (191, 192) have provided the bulk of theindirect evidence supporting the hypothesis that genomic instability is a potential mechanism driving tumor progression. Theevidence is less compelling (193) when the frequencies of eitherspontaneous point mutations (194-201) or drug-induced geneamplification (202-206) are compared in normal and neoplasticcells.

Recombinase infidelity (207), telomere reduction (208), andDNA hypomethylation (47, 209) are examples of additionalmechanisms of genomic instability. The frequency of somaticrecombinational events at certain loci, e.g., ¡mmunoglobulingenes, is relatively high, but specific DNA sequences at theseloci are recognized by the responsible V-(D)-J recombinases.Although the degree of infidelity of wild type and mutantrecombinases is not known, a possible example of "illegitimate"

5031s

on April 2, 2020. © 1991 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Page 10: Chemical and Physical Carcinogenesis: Advances …...CARCINOGENESIS Table 1 Examples of advances in chemical and physical carcinogenesis" A. Cancer etiology I. Life-style factors (tobacco

CARCINOGENESIS

Table 6 Examples of gene abnormalities associated with genomic instability

Genomic inslabilily

GeneRAD

9RAD18p}4«"Topoisomerase

IInodDNA

polymerase«p53R.4C-IseidUnknownSpeciesS.

cererisiaeS.cerevisiae.9.pombeS.pombeDrosophilaHumanHuman

(Li-Fraumenisyndrome)HumanMurineHuman

(Bloom's syndrome)ActivationMutationMutationMutationMutationMutationMutationMutationMutation"MutationUnknownTypeChromosomal

lossBasesubstitution (G:C—>T:Atransversion)AneuploidyAbnormal

chromosomalsegregationAbnormalchromosomalsegregationBase

substitutionAneuploidyChromosomal

rearrangements anddeletionsAberrantt'(D)J recombination anddouble-strandbreak

DNArepairHyperrecombination

' In vitro experimental studies.

V-(D)-J recombinase activity leading to interstitial deletion andrearrangements in a growth-regulatory gene has been described(210). In addition, the seid (severe combined immunodeficiency)mutation in mice is due to a defect in the site-specific V(D)Jrecombination pathway and has been recently linked to a deficiency in double-strand break DNA repair. This linkage suggests that the seid gene product performs a similar function inboth pathways.

Hastie et al. (208) have shown that telomere reduction iscommon in both colorectal adenomas and carcinomas and havespeculated that telomere reduction may be involved as an earlyevent in carcinogenesis. Interestingly, studies of yeast (211) andmaize (212) have shown that chromosomes without terminalrepeats or telomeres are prone to rearrangements or loss. Studies in yeast have also identified genes involved in cell cyclecontrol and mitosis, e.g., RAD9 and cdc-2, that, when mutated,influence the fidelity of mitosis (213, 214) (Table 6). Forexample, certain mutations in RAD9 of 5. cerevisiae increasethe rate of chromosomal loss by 7-21-fold (215) and topoisom-erase II mutants in Saccharomyces pombe have abnormal segregation of the intertwined daughter chromosomes at the endof DNA replication (216) and abnormal chromosome condensation (217). Since human homologues of such genes are beingidentified, similar investigations could be conducted using cultured human cells, and human tumors could be examined formutations in these genes.

Analogous to the yeast paradigm of a mutator phenotypeassociated with dysregulation of cell cycle control (213), certainactivated protooncogenes or inactivated tumor suppressorgenes could lead to genomic instability. The rapid appearanceof aneuploidy and "spontaneous" immortalization of cultured

human fibroblasts from donors with the Li-Fraumeni syndrome, who have an inherited mutant p53 alíele(218), isconsistent with this hypothesis. The genomic instability causedby certain DNA viruses such as SV40, adenovirus, and papil-lomavirus could also be due to the inactivation of p53 and/orRb by complexing with the viral proteins.

As argued by Loeb (55), the impression of a high degree ofgenomic stability may be misleading when one considers thatthe human body contains about IO14cells and that, within theaverage human life span, the cells undergo a total of IO'6

divisions. Therefore, spontaneous mutagenesis may significantly contribute to the prevalence of certain cancer types thathave no apparent risk factors and minimal variations in incidence occur among different geographic areas. However, epidemiológica! studies have: (a) identified significant cancer riskfactors, e.g., tobacco smoke and hepatitis B virus; (b) foundsubstantial geographic variation in the incidence of the mostcommon types of human cancer; and (c) found that migrant

populations frequently acquire the risk of cancer prevailing intheir new geographic location of residence (1, 2, 219). Basedon the above and other considerations ( 1), it has been estimatedthat about 80% of all cancer in the United States is due toenvironmental factors. Finally, one can speculate that the ratesof spontaneous mutations, e.g., deamination of 5-methylcyto-sine, may be altered by environmental chemicals directly and/or indirectly via generation of endogenous agents such as oxyradicals and the intracellular signal transducer, nitric oxide.

Molecular Epidemiology of Human Cancer

Combined laboratory-epidemiological studies of human cancer have a long history and have been named metabolic (220),biochemical (221), and molecular epidemiology (222). Whereasclassical epidemiology has been successful in identifying highcancer risk populations, e.g., cigarette smokers, the primarygoal of molecular epidemiology is to identify individuals inthese populations who are at the highest cancer risk. Thecurrent strategy involves improved exposure assessment bymeasuring biologically effective doses of carcinogens and secondly, analysis of host susceptibility factors within the framework of well-designed epidemiological studies (223).

Exposure Assessment

Carcinogen-macromolecular adducts, mutations, and cyto-genetic changes can be measured in the target epithelial cellpopulation and in surrogate blood cells as shown in Fig. 3. Thisparadigm is now feasible to explore experimentally because ofrecent advances in methods to measure carcinogen-macromo-lecular adducts (224), somatic gene mutations (181, 225), andcytogenetic endpoints (103). In addition, polymerase chainreaction technology and DNA sequencing allows a rapid assessment of mutations in protooncogenes, tumor suppressorgenes, and antimetastasis genes in the epithelial cells. Since theras protooncogene family is occasionally and the p53 tumorsuppressor gene is frequently mutated in human cancers, asdiscussed above, these genes are suitable candidates for assessing the genetic consequences of carcinogen-DNA adducts andendogenous mutagens. The mutational spectra in ras and p53genes can be measured by current technology due to the clonalexpansion of the cells driven by these mutant genes. Althoughsmall clonal populations of a few hundred cells are sufficientfor analysis of mutational spectra by polymerase chain reactionand direct DNA sequencing, additional advances in methodology will be required to determine the mutational spectra insingle cells that have not clonally expanded and are diluted ina much larger population of normal cells.

5032s

on April 2, 2020. © 1991 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Page 11: Chemical and Physical Carcinogenesis: Advances …...CARCINOGENESIS Table 1 Examples of advances in chemical and physical carcinogenesis" A. Cancer etiology I. Life-style factors (tobacco

CARCINOGENESIS

Host Susceptibility Factors

Both inherited and acquired host factors that predispose anindividual to cancer caused by environmental agents have beenidentified (37, 226). Inheritance of a germline mutation in thetumor suppressor genes, p53 (141, 142) or, perhaps, Rb (227),predisposes to the development of adult types of cancer (Table7). Other inherited host susceptibility factors include certainDNA repair deficiencies (228), genetic polymorphism in certainxenobiotic metabolizing enzymes (70), and, perhaps, the proox-idant state in Bloom's syndrome (229).

Interindividual Variation in Human Carcinogenesis

No one supposes that all the individuals of the same species arecast in the very same mould. These individual differences arehighly important for us. ...

The Origin of SpeciesCharles Darwin, 1859

Scientists have repeatedly recognized person-to-person differences in behavior, morphology, and risk of disease. Suchinterindividual differences may be either inherited or acquired.For example, inherited differences in susceptibility to physicalor chemical carcinogens have been observed among individuals,including an increased risk of sunlight-induced skin cancer inpeople with xeroderma pigmentosum (228), bladder cancer indye stuff workers with a poor acetylator phenotype (230), andbronchogenic carcinoma in tobacco smokers who have an extensive debrisoquine hydroxylator phenotype (231, 232).

Because most chemical carcinogens require metabolic activation to exert their oncogenic effects and the amount ofultimate carcinogen produced results from the action of competing activation and detoxication pathways, interindividualvariation in carcinogen metabolism is considered to be animportant determinant of cancer susceptibility (75). DNA ad-ducts are one form of genetic damage caused by chemicalcarcinogens and may lead to mutations that activate protoon-

•Environmental Exposure

J

Table 7 Examples of inherited predisposition of human cancer

1•Bio1

rbcsei

ir

odI•um

wbcr

*•AdductsG.A.•

CytogeneticEndpoints•«

•^CellClonal

Expansion•

Mutationalspectra, e.g.,HPRT, HLA-*n•

Targetf Tissue(cell)»•

•Adducts1^

•CytogeneticEndpointsCellDonai

Expansion•

Mutational— *• spectra, e.g.,

ras, p53

•Exposure Assessment

Fig. Õ.Molecular dosimetry, mutational spectra, and exposure assessment.Paradigm for improvement of carcinogen exposure assessment that involves themeasurement of carcinogen-macromolecular adducts, cytogenetic end points, andmutational spectra in protooncogenes, e.g., ras. or tumor suppressor genes, e.g.,p53, found in target tissues and in monitoring genes, e.g., hprt or histocompati-bility genes (HLA), in blood lymphocytes or glycophorin A (G.A.) protein in redblood cells (rbc).

CandidategenesP53RbMCCCYP2D6XPACConditionLi-Fraumeni

syndromeFamilial

retinoblastomaFamilialpolyposiscoliExtensive

debrisoquine phenotypeXeroderma

pigmentosum AExamples

ofcancertypeCarcinomas

ofthebreastand blad

der;sarcomasRetinoblastomaColonLungSkin

cogenes and inactivate tumor suppressor genes in replicatingcells. The steady state levels of these adducts depend on boththe amount of ultimate carcinogen available to bind and therate of removal from DNA by enzymatic repair processes. Thegenomic distribution of adduct formation and repair is nonran-dom and is influenced by both DNA sequence and chromatinstructure (121, 233-235).

Carcinogen Metabolism and DNA Adduct Formation. Chemical carcinogens are metabolized by a wide variety of solubleand membrane-bound enzymes. Multiple forms of human cy-tochrome P-450 are involved in the oxidative metabolism ofchemical carcinogens, e.g., polycyclic aromatic hydrocarbons(70). Several thousand-fold interindividual variation has beenobserved in placental AHH activity which is catalyzed by theCYPIA1; some of this variability is under direct genetic control,but variations are also the result of an enzyme induction processdue to maternal exposure to environmental carcinogens, e.g.,tobacco smoke, or dietary factors (236-238). However, theinduction process itself may have a genetic component (239)and inducible AHH activity is higher in cultured lymphocytesfrom lung cancer cases when compared to controls (240). Trelland coworkers (241) are currently conducting a prospectivestudy to determine if high inducibility of AHH is a risk factorfor the development of cancer. Increased binding ofbenzo(a)pyrene metabolites to DNA in lymphocytes of patientswith lung cancer versus noncancerous controls has also beenobserved-(242). A highly inducible allelic variant CYPIA1 hasbeen identified in humans (243).9 This variant is associatedwith increased risk of smoking-associated lung cancer in aJapanese population (244). This association has not been foundin a U.S. population10.

Cytochrome P-450 (CYP2D6) activity is polymorphic andhas also been linked to lung cancer risk (231, 232). CYP2D6hydroxylates xenobiotics such as debrisoquine, an antihyperten-sive drug, and a tobacco-specific A'-nitrosamine (70), and anindividual's polymorphic phenotype is inherited in an autoso-

mal recessive manner. The rate of 4-hydroxylation of debrisoquine varies several thousand-fold among people, and lung,liver, or advanced bladder cancer patients are more likely tohave the extensive hydroxylator phenotype when compared tononcancer controls (231, 245, 246). In a case-control study oflung cancer in the United States, the extensive hydroxylatorphenotype had an 8-fold increased cancer risk when comparedto the poor hydroxylator phenotype and the increased risk wasprimarily for histológica! types other than adenocarcinoma ofthe lung (232). In addition, analysis of data derived from aBritish population (231) has revealed that individuals with theextensive hydroxylator phenotype who are occupationally ex-

9T. L. McLemore, S. Adelberg, and M. C. Liu. Cytochrome P450IA1 geneexpression in lung cancer patients: evidence for cigarette smoke-induced expression in normal lung and altered gene regulation in primary pulmonary carcinomas,submitted for publication.

IOP.Shields, A. Weston. and C.C. Harris, unpublished results.

5033s

on April 2, 2020. © 1991 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Page 12: Chemical and Physical Carcinogenesis: Advances …...CARCINOGENESIS Table 1 Examples of advances in chemical and physical carcinogenesis" A. Cancer etiology I. Life-style factors (tobacco

CARCINOGENESIS

posed to high amounts of either asbestos or polycyclic aromatichydrocarbons have relative lung cancer risks of 18- and 35-fold,respectively (247). CYP2D6 may activate a chemical carcino-gen(s) found in tobacco smoke, e.g., certain jY-nitrosamines,and/or, a more speculative possibility, inactivate nicotine, theaddictive pleasure component of tobacco smoke that woulddecrease its steadystate level and increase the drive to smokemore cigarettes. Thereby, the individual with the extensivemetabolizer phenotype would be at greater cancer risk. Anotherhypothesis is that an alíeleof the CYP2D6 gene is in linkagedisequilibrium with another gene that influences cancersusceptibility.

The ¿Y-acetylationpolymorphism is controlled by two auto-somal alíelesat a single locus in which rapid acetylation is thedominant trait and slow acetylation is recessive. Acetylation ofcarcinogenic aromatic amines has been proposed as a cancerrisk factor (248). The slow acetylator phenotype has been linkedto occupationally induced bladder cancer in dye workers exposed to large amounts of A'-substituted aryl compounds (230).

Interestingly, the rapid acetylator phenotype is commonlyfound in colon cancer cases (249). Whether or not this association is due to metabolism of a carcinogenic aromatic amine inthe colonie epithelium is not known.

Wide interindividual differences in detoxifying enzymes ofcarcinogens are also found. For example, at each step in themetabolic pathway of benzo(a)pyrene activation to electrophilicdiol epoxides, competing detoxifying enzymes are found (250).Confirming previous observations (251), a recent study of several of the enzymes involved in benzo(a)pyrene metabolismshowed a more than 10-fold person-to-person variation in theiractivities (252) and presented indirect evidence that tobaccosmoke induced many of these enzymes. Genetic control of thepresumed detoxication of benzo(a)pyrene by conversion towater-soluble metabolites has been reported (239).

GSTs are multifunctional proteins (253) that catalyze theconjugation of glutathione to electrophiles, including the ultimate carcinogenic metabolite of benzo(a)pyrene (254), and areconsidered to be a detoxification pathway of carcinogenic poly-cyclic aromatic hydrocarbons (255). The three isoenzymes ofGST («-<,n and w) vary in their substrate specificity, tissuedistribution, and activities among individuals (256). Expressionof GST-ji is inherited as an autosomal dominant trait (257) andindividuals with low GST-^ activity may be at greater risk oflung cancer caused by cigarette smoking (258, 259). Furtherfunctional and genetic studies of acquired and inherited deficiencies in the activities of enzymes involved in the detoxification of chemical carcinogens are warranted.

Metabolism of carcinogens from several chemical classes,including /V-nitrosamines, polycyclic aromatic hydrocarbons,hydrazines, mycotoxins, and aromatic amines, has been studiedusing human tissues, cells, and microsomes (73, 260-267). Theenzymes responsible for the activation and deactivation ofprocarcinogens, the metabolites produced, and the carcinogen-DNA adducts formed by cultured human tissues and cells, ingeneral, are qualitatively similar among donors and tissue types.The DNA adducts and carcinogen metabolites are also similarto those found in most laboratory animals, an observation thatgenerally supports the qualitative extrapolation of carcinogen-esis data from the laboratory animal to the human (73). However, some notable differences among animal species have beenreported, including metabolism of aromatic amines in theguinea pig (65), benzo(a)pyrene in the rat and rabbit (66, 268),and aflatoxin B, in the Syrian golden hamster (67).

Although the major DNA adducts are qualitatively similarfor the chemical carcinogens thus far studied in these in vitromodels, quantitative differences have been found among individuals and their various tissue types (73, 260, 269-271). Thedifferences in formation of DNA adducts range from approximately 10-150-fold among humans, the interindividual distribution is generally unimodal, and the variation is similar inmagnitude to that found in pharmacogenetic studies of drugmetabolism (75).

DNA Repair Rates. DNA repair enzymes modify DNA damage caused by carcinogens in reactions that generally result inthe removal of DNA adducts. Studies of cells from donors withxeroderma pigmentosum have been particularly important inexpanding our understanding of DNA excision repair and itspossible relationship to risk of cancer (228). The rate but notthe fidelity of DNA repair can be determined by measuringunscheduled DNA synthesis and removal of DNA adducts, andsubstantial interindividual variations in DNA repair rates havebeen observed (272). As discussed above, the fidelity of DNArepair could also vary among individuals, and recent advancesin the identification of mammalian DNA repair genes and theirmolecular mechanisms (273, 274) should provide an opportunity to investigate the fidelity of excision DNA repair in thenear future. In addition to finding excision repair rates severelydepressed in xeroderma pigmentosum cells (e.g., complementation group A), an approximately 5-fold variation amongindividuals in unscheduled DNA synthesis induced by UVexposure of lymphocytes in vitro has been found in the generalpopulation (272). A significant reduction in unscheduled DNAsynthesis induced in vitro by yV-acetoxy-2-acetylaminofluorenehas been observed in mononuclear leukocytes from individualswith a history of cancer in first degree relatives when comparedto those without a family history (275, 276).

Interindividual variation has been noted in the activity of O6-alkyldeoxyguanine-DNA alkyltransferase; this enzyme repairsalkylation damage to O6-deoxyguanine (277-280). In additionto these person-to-person differences of about 40-fold, widevariations in this DNA repair activity have been observed indifferent types of tissues (277, 278, 281, 282), fetal tissuesexhibit 2- to 5-fold less activity than the corresponding adulttissues (283), and cells may have lower repair rates after terminal differentiation (284). The activity of this DNA repairenzyme is inhibited by certain aldehydes (281) and alkylatingcancer chemotherapeutic agents (285). A decrease in this DNArepair activity has been observed in fibroblasts from patientswith lung cancer when compared to donors with either melanoma or noncancer controls (286). Therefore, acquired and/orinherited deficiency in 06-alkyldeoxyguanine-DNA alkyltrans

ferase may be a cancer risk factor in tobacco smokers.A unimodal distribution of repair rates of benzo(a)pyrene

diol-epoxide-DNA adducts has been observed using humanlymphocytes in vitro (287). The interindividual variation wassubstantially greater than the intraindividual variation whichsuggests a role of inherited factors. The influence of thesevariations in DNA repair rates in determining tissue site andrisk of cancer in the general population remains to bedetermined.

Tumor Promotion. Because cancer is the result of complexinteraction between multiple environmental factors and bothacquired and inherited host factors, one should consider carcin-ogen-DNA adducts as only one piece in the puzzle. Other piecesinclude determinants of tumor promotion. In the skin carcino-genesis studies, wide variations in susceptibility to tumor-pro-

5034s

on April 2, 2020. © 1991 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Page 13: Chemical and Physical Carcinogenesis: Advances …...CARCINOGENESIS Table 1 Examples of advances in chemical and physical carcinogenesis" A. Cancer etiology I. Life-style factors (tobacco

CARCINOGENFSIS

moling agents have been observed among animal species andamong different inbred strains of a single species (288-291).Epidemiological studies suggest that tumor promotion mayinfluence both tumor probability and latency period in humans(1), and mathematical models of carcinogenesis indicate thatcell proliferation is an important determinant of cancer risk(292). Methods to identify human tumor promoters and topredict responses to tumor promoters among different humansneed to be developed.

Interindividual Variation in Carcinogen-Macromolecular Ad-ducts in People Exposed to Chemical Carcinogens. Results fromin vivo and in vitro studies discussed above serve as a basis forinvestigations in biochemical and molecular epidemiology. Forexample, the observation that the carcinogen-DNA adductsformed in cultured human tissues are generally the same asthose found in experimental animals in which these chemicalsinduce cancer has encouraged investigators to search- for DNA

adducts in biological specimens obtained from people exposedto either specific carcinogens such as benzo(a)pyrene or chem-

otherapeutic agents.The development of highly sensitive methods to detect car-

cinogen-macromolecular adducts has made it possible to measure adducts in blood and tissue samples (293-295). Althoughhemoglobin and albumin are not considered targets for thepathobiological effects of carcinogens, these macromoleculeshave certain advantages for the molecular dosimetry of exposure to carcinogens. For example, RBC have a lifetime of about120 days in humans, so the levels of carcinogen-hemoglobinadducts may be an integrative measure of exposure during aperiod of nearly 4 months. In addition, these protein adducts,unlike DNA adducts, are not considered to be repaired in thecirculating RBC, and mg quantities of hemoglobin can beobtained from a few ml of blood. Ehrenberg et al. (296) pioneered this area of research by measuring alkylating agentssuch as ethylene oxide bound to hemoglobin. The amounts ofethylene oxide-hemoglobin and 4-aminobiphenyl-hemoglobinadducts are substantially increased in tobacco smokers compared to nonsmokers (297, 298), and the 4-aminobiphenyl-hemoglobin adducts levels decrease upon smoking cessation(299). The kinetics of adduci loss mirrors the 120-day half-lifeof the erythrocytes. Occupational exposure lo elhylene oxidehas also been monilored (300). Dietary exposure to chemicalcarcinogens can be monitored by measuring carcinogen-albumin adducts. Wild et al. (301) have found thai exposure toaflaloxin B, in coniaminaled grain products is widespread inThe Gambia and Senegal, >95% of the subjects had deteclableaflatoxin-albumin adducts, and the interindividual variationeven within a single village can be 100-fold.

Carcinogen-DNA adducts have also been measured in workers exposed to chemical carcinogens usually in complex mixtures such as coke oven exhaust. For example, PAH-DNAadducts in peripheral blood lymphocytes have been measuredby enzyme immunoassays in coke oven workers, foundry workers, roofers, and fire fighters (302-307). In the occupationallyexposed individuals, enzyme immunoassays detecled 1 to 1200adducts (BPDE-DNA aniigenic equivalent) per IO8 unmodified nucleolides in 25 to 100% of the subjects (303-305, 307-309). These adducts have also been detected by complementarymethods in placenta! DNA samples in levels that ranged between 10 and 500 in 10s nucleotides. In this case, synchronous

scanning fluorimetry and gas chromalography-mass spectros-copy have given consistent results (310-315).

A comparison between deteclion methods and end poinls has

also been performed for coke oven workers in ihe United Statesand Scandinavian countries (308, 316). However, levels ofPAH-DNA adducts reported using the 12P-postlabeling assay

for detection are lower than those reported using enzyme im-munoassay for deteclion. These dala can in pari be explainedby ihe relalive subslrate specificities of each assay which havenot yet been fully slandardized or rigorously compared. Anli-PAH-DNA anlibodies that presumably indicate genotoxic exposure to metabolically aclivaled PAHs have also been detectedin human serum of both occupalionally exposed and nonex-posed populations (coke oven workers; smokers and non-smokers) (303, 304, 309).

Alkyl-DNA adducts have also been detecled in human samples. The receñíuse of HPLC or ¡mmunoaffinily chromatog-raphy in combination with the 32P-postlabeling assay has revealed Ihe presence of 06-methyldeoxyguanosine, O6-ethyl-deoxyguanosine, O4-ethylthymidine, and 7V7-melhyldeoxy-

guanosine as well as other types of alkyl adducls in ihe humanpopulation, including cancer patients (317-322). The combination of HPLC and the "P-posllabeling assay has been usedto detect O6mdG in lung cancer cases (320, 323) and immu-

noaffmily chromalography and HPLC have been used lo deleclIhese alkyl adducts in placenta! DNA (321, 324). The power ofHPLC combined with the "P-posllabeling assay is evidenlbecause O6-elhyldeoxyguanosine and /V7-methyldeoxyguano-

sine adducts can be detected with a high degree of sensitivityand specificily in lung lissues al the level of 1 adduci in IO7

nucleotides (325, 326).The wide range in the amounts of carcinogen-macromolecu-

lar adducls measured in ihe sludies discussed above is in pari areflection of occupational, environmenl, or life-style exposurelo chemical carcinogens. Inlerindividual differences in ihe balance belween metabolic activation and deactivation of chemicalcarcinogens and in DNA repair rates are also likely to conlrib-ute to the variation.

Although methods to measure carcinogen-macromolecularadducts are an importanl conlribulion to improved carcinogenexposure assessment, carcinogen-macromolecular adducls willnoi be a precise quanlitative predictor of cancer risk because ofthe multistage and complex nalure of human carcinogenesis.

In biology, one rarely deals with classes of identical entities, butnearly always studies populations consisting of unique individuals.This is true for every level of hierarchy, from cells to ecosystems.

The Growth of Biological ThoughtErnst Mayr, 1982

Cancer Risk Assessment Perspective

Quantitative risk assessment is currenlly more mathematicalextrapolation and scientific inluition than a rigorous science.However, the stalus of cancer risk assessmenl should improveduring the 1990s. This optimism is based largely on the recentadvances that have increased our understanding of the molecular mechanisms of human disease palhogenesis. The discoveryof multiple genetic and epigenelic changes during carcinogenesis is an example lhal provides opporlunilies for bolh earlydiagnosis of preneoplaslic lesions and novel interventive methods to inhibit or reverse carcinogenesis. These advances aredriven in pari by improved biotechnology. For example, polym-erase chain reaction coupled wilh direcl DNA sequencing allows the detection of mutalions in formalin-fixed, paraffin-embedded tissue sections and the "retrospective epidemiologi-

5035s

on April 2, 2020. © 1991 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Page 14: Chemical and Physical Carcinogenesis: Advances …...CARCINOGENESIS Table 1 Examples of advances in chemical and physical carcinogenesis" A. Cancer etiology I. Life-style factors (tobacco

CARCINOGENESIS

cal" study of archival material (327).

Quantitative methods to measure human exposure to biologically effective doses of chemical and physical carcinogenscontinue to improve and their current limitations and advantages have been extensively discussed (224,293). If developmentand validation continue at the present pace, methodology forcarcinogen exposure assessment should be satisfactory by theend of this decade.

The identification of inherited or acquired host susceptibilityfactors is also becoming an important factor in the risk assessment equation (223) (Table 7). Analogous to the confoundmentof exposure assessment by complex mixtures, time course variables, etc., generic problems concerning the use of data on hostsusceptibility factors in risk assessment are evident: (a) rankorder of "potency or weight" of various host susceptibility

factors influencing the cancer risk in individuals exposed tocarcinogens; (b) "potency" impact of the degree of the interin

dividual variation in a specific host susceptibility factor; and (c)interactive effects among various host susceptibility factors. Inaddition, societal and ethical issues come to the forefront whenever individuals at high disease risk are identified. These issuesare already being actively discussed (328). Finally, the conductof well-designed and multifaceted molecular epidemiológica!studies will require the development of a new cadre of scientistswho have knowledge and skills in both laboratory science andanalytical epidemiology.

By the end of the 1990s, molecular epidemiology of humancancer risk is predicted to have a substantial impact on thequality of quantitative risk assessment and should contribute tothe identification of individuals who will most benefit by cancerprevention strategies including intervention by chemopreven-tive therapy during the multistage process of carcinogenesis.

Acknowledgments

The editorial aid of Robert Julia is appreciated. The comments ofMarshal Anderson, Allan Conney, John Essigmann, Frank Gonzalez,John Higginson, Lawrence Loeb, James Miller, Janardan Reddy, Phillip Shubik, Gary Stoner, Benjamin Trump, Bert Vogelstein, ElizabethWeisburger, Ainsley Weston, Gerald Wogan, and Stuart Yuspa areappreciated.

References

1. Doll. R., and Peto. R. The Causes of Cancer. New York: Oxford Press,1981.

2. Muir. C. S. Epidemiology, basic science, and the prevention of cancer:implications for the future. Cancer Res.. 50: 6441-6448. 1990.

3. Fraumeni, J. F., Jr., Hoover, R. N., Devesa, S. S., and Kinlen, L. J.Epidemiology of cancer. In: V. T. DeVita, Jr., S. Hellman, and S. A.Rosenberg (eds.). Cancer. Principles and Practice of Oncology, pp. 196-235. Philadelphia: J. B. Lippincott Co.. 1990.

4. Yuspa, S. H.. and Poirier. M. C. Chemical carcinogenesis: from animalmodels to molecular models in one decade. Adv. Cancer Res., 50: 25-70.1988.

5. Heidelberger, C. Chemical oncogenesis in culture. Adv. Cancer Res., IS:317-366, 1973.

6. Harris, C. C. Human tissues and cells in carcinogenesis research. CancerRes., 47: 1-10. 1987.

7. Bishop. J. M. The molecular genetics of cancer. Science (Washington DC).235: 305-311. 1987.

8. Klein. G. Oncogenes and tumor suppressor genes. Acta Oncol., 27: 427-437, 1988.

9. Weinberg, R. A. Oncogenes, antioncogenes. and the molecular bases ofmultistep carcinogenesis. Cancer Res., 49: 3713-3721. 1989.

10. Harris. H. The role of differentiation in the suppression of malignancy. J.Cell Sci., 97:5-10, 1990.

11. Stanbridge, E. J. Human tumor suppressor genes. Annu. Rev. Genet.. 24:615-657, 1990.

12. Howley, P. M. The role of the human papillomav¡ruses in human cancer.Cancer Res.. 5/(Suppl.): 5019s-5022s, 1991.

13. Searle, C. E. Chemical Carcinogens. ACS Monograph 182. Washington.DC: American Chemical Society. 1984.

14. Farber, E. The multistep nature of cancer development. Cancer Res., 44:4217-4223. 1984.

15. Hiatt, H.H.. Watson, J. D., and Winsten, J. A. Origins of Human Cancer.Book B: Mechanisms of Carcinogenesis. Cold Spring Harbor, NY: ColdSpring Harbor Laboratory Press, 1977.

16. Essex, M., Todaro, G., and zur Hausen. H. Viruses in Naturally OccurringCancers. Book A. Cold Spring Harbor. NY: Cold Spring Harbor LaboratoryPress, 1980.

17. Pilot, H. C. Principles of carcinogenesis: chemical. In: V. T. DeVita, Jr., S.Hellman, and S. A. Rosenberg (eds.). Cancer. Principles and Practice ofOncology, pp. 116-135. Philadelphia: J. B. Lippincott Co.. 1990.

18. Fry, R. J. M. Principles of carcinogenesis: physical. In: V. T. DeVita, Jr..S. Hellman. and S. A. Rosenberg (eds.). Cancer. Principles and Practice ofOncology, pp. 136-148. Philadelphia: J. B. Lippincott Co.. 1990.

19. Bishop, J. M. Molecular themes in oncogenesis. Cell, 64: 235-248, 1991.20. Hunter, T. Cooperation between oncogenes. Cell, 64: 249-270, 1991.21. Marshall, C. Tumor suppressor genes. Cell, 64: 313-326. 1991.22. Weston. A., and Harris. C. C. Chemical carcinogenesis. In: J. F. Holland.

E. Frei, III, R. C. Bast. Jr.. D. W. Kufe. D. L. Morton, and R. R.Weichselbaum (eds.). Cancer Medicine. Ed. 3. Philadelphia: Lea & Febiger,in press, 1991.

23. Weisburger, J. H., and Williams, G. M. Chemical Carcinogenesis. In: J. F.Holland and E. Frei (eds.). Cancer Medicine, Ed. 2, pp. 42-95. Philadelphia:Lea and Febiger, 1982.

24. Harris, C. C., and Autrup. H. N. Human Carcinogenesis. New York:Academic Press. 1983.

25. Brugge. J.. Curran, T.. Harlow. E., and McCormick, F. Origins of HumanCancer. Cold Spring Harbor, NY: Cold Spring Harbor Laboratory Press,1991.

26. Yuspa, S. H., and Harris, C. C. Molecular and cellular basis of chemicalcarcinogenesis. In: D. Schottenfeld and J. F. Fraumeni (eds.). CancerEpidemiology and Prevention, pp. 23-43. Philadelphia: W. B. SaundersCo., 1982.

27. Cerutti. P. A. Prooxidant states and tumor promotion. Science (WashingtonDC). 227:375-381, 1985.

28. Weinstein, I. B.. Borner. C. M., Krauss, R. S., OTJriscoll. K., Choi, P. M.,Moritomi, M., Hoshina, S., Hsieh, L. L.. Tchou-Wong, K. M., Guadagno,S. N., Ueffing. M., and Guillem. J. Pleiotropic effects of protein kinase Cand the concept of carcinogenesis as a progressive disorder in signal trans-duction. In: J. Brugge. T. Curren. E. Harlow. and F. McCormick (eds.).Origins of Human Cancer. New Y'ork: Cold Spring Harbor Laboratory

Press, in press. 1991.29. Willey, J. C, Moser, C. E., Jr., Lechner. J. F., and Harris, C. C. Differential

effects of 12-O-tetradecanoylphorbol-13-acetate on cultured normal andneoplastic human bronchial epithelial cells. Cancer Res., 44: 5124-5126,1984.

30. Parkinson, E. K. Defective responses of transformed keratinocytes to terminal differentiation stimuli. Their role in epidermal tumour promotion byphorbol esters and by deep skin wounding. Br. J. Cancer. 52: 479-493,1985.

31. Pfeifer, A., Lechner, J. F., Masui, T., Reddel. R. R., Mark. G. E., andHarris, C. C. Control of growth and squamous differentiation in normalhuman bronchial epithelial cells by chemical and biological modifiers andtransferred genes. Environ. Health Perspect., 80: 209-220. 1989.

32. Yuspa, S. H.. Ben, T.. Hennings. H., and I nini LI. Divergent responses inepidermal basal cells exposed to the tumor promoter 12-0-tetradecanoyl-phorbol-13-acetate. Cancer Res., 42: 2344-2349, 1982.

33. Parkinson, E. K., Grabham, P., and Emmerson, A. A subpopulation ofcultured human keratinocytes which is resistant to the induction of terminaldifferentiation—related changes by phorbol, 12-myristate, 13-acetate: evidence for an increase in the resistant population following transformation.Carcinogenesis (Lond.). 4: 857-861. 1983.

34. Moses, H. L., Y'ang, E. Y., and Pietenpol, J. A. TGF-tf stimulation and

inhibition of cell proliferation: new mechanistic insights. Cell, 63: 245-247,1990.

35. Roteilo, R. J.. Lieberman, R. C., Purchio, A. F.. and Gerschenson. L. E.Coordinated regulation of apoptosis and cell proliferation by transforminggrowth factor beta 1 in cultured uterine epithelial cells. Proc. Nati. Acad.Sci. USA. 88: 3412-3415. 1991.

36. Slaga. T. J. Overview of tumor promotion in animals. Environ. HealthPerspect., 50:3-14, 1983.

37. Haugen, A., and Harris, C. C. Interactive effects between viruses andchemical carcinogens. In: C. S. Cooper and P. L. Grovcr (eds.). Carcinogenesis and Mutagenesis. Handbook of Experimental Pharmacology, pp. 249-268. London: Springer-Verlag, 1990.

38. Hennings, H.. Shores, R., Wenk. M. L.. Spangler, E. F., Tarone. R., andYuspa, S. H. Malignant conversion of mouse skin tumours is increased bytumour initiators and unaffected by tumour promoters. Nature (Lond.), 304:67-69. 1983.

39. O'Connell. J. F., Klein-Szanto, A. J.. Digiovanni, D. M.. Fries, J. W., and

Slaga. T. J. Malignant progression of mouse skin papillomas treated withethylnitrosourea, A'-methyl-A"-nitro-A'-nitrosoguanidine, or 12-O-tetra-

decanoylphorbol- 13-acetate. Cancer Lett.. 30: 269-274, 1986.40. Hennings, H. Malignant conversion: the first stage in progression from

benign to malignant tumors. In: T. J. Slaga, A. J. P. Klein-Szanto, R. K.

5036s

on April 2, 2020. © 1991 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Page 15: Chemical and Physical Carcinogenesis: Advances …...CARCINOGENESIS Table 1 Examples of advances in chemical and physical carcinogenesis" A. Cancer etiology I. Life-style factors (tobacco

CARCINOGENESIS

Boutwell, D. E. Stevenson, H. L. Spitzer, and B. D'Motto (eds.). SkinCarcinogenesis, pp. 95-105. New York: Alan R. Liss, Inc.. 1989.

41. Yuspa. S. H., Hennings, H., Roop, D., Strickland, J., and Greenhalgh, D.A. Genes and mechanisms involved in malignant conversion. In: C. C. 73.Harris and L. A. Liotta (eds.). Genetic Mechanisms in Carcinogenesis andTumor Progression, pp. 115-126. New York: Wiley-Liss, 1990.

42. Foulds, L. The experimental study of tumor progression: a review. Cancer 74.Res., 14: 317-339, 1954.

43. Nowell. P. C. The cioncai evolution of tumor cell populations. Science(Washington DC), 194: 23-28. 1976.

44. Rowley. J. D. Molecular cytogenetics: Rosetta stone for understandingcancer—twenty-ninth G. H. A. Clowes Memorial Award Lecture. Cancer 75.Res., 50:3816-3825, 1990.

45. Schimke, R. T. The search for early genetic events in tumorigenesis: anamplification paradigm. Cancer Cells (Cold Spring Harbor), 2: 149-151, 76.1990.

46. Peto. R., Roe, F. J., Lee, P. N., Levy, L., and Clack, J. Cancer and ageingin mice and men. Br. J. Cancer. 32: 411-426, 1975.

47. Fearon, E. R., and Vogelstein. B. A genetic model for colorectal tumorigenesis. Cell, 61: 759-767, 1990. 77.

48. Cairns. J. Mutation selection and the natural history of cancer. Nature(Lond.), 255: 197-200, 1975.

49. Butterworth. B. E.. and Slaga. T. J. Nongenotoxic Mechanisms in Carci- 78.nogenesis. Banbury Report Vol. 25. Cold Spring Harbor. NY: Cold SpringHarbor Laboratory Press, 1987.

50. Wilson, V. L., Smith, R. A., Longoria. J., Liotta, M. A., Harper, C. M.,and Harris, C. C. Chemical carcinogen-induced decreases in genomic 5- 79.methyldeoxycytidine content of normal human bronchial epithelial cells.Proc. Nati. Acad. Sci. USA, 84: 3298-3301. 1987.

51. Lutz. W. K. Dose-response relationship and low dose extrapolation inchemical carcinogenesis. Carcinogenesis (Lond.), //: 1243-1247, 1990. 80.

52. Swenberg. J. A.. Richardson. F. C.. Boucheron, J. A., Deal, F. H., Belinsky.S. A., Charbonneau, M., and Short, B. G. High- to low-dose extrapolation:critical determinants involved in the dose response of carcinogenic substances. Environ. Health Perspect., 76: 57-63. 1987. 81.

53. Hoel, D. G., Haseman. J. K., Hogan, M. D., Huff, J., and McConnell, E.E. The impact of toxicity on carcinogenicity studies: implications for riskassessment. Carcinogenesis (Lond.), 9: 2045-2052, 1988.

54. Ames. B. N. Endogenous DNA damage as related to cancer and aging. 82.Mutât.Res., 214: 41-46, 1989.

55. Loeb, L. A. Mutator phenotype may be required for multistage carcinogenesis. Cancer Res., 51: 3075-3079, 1991.

56. Lutz, W. K. Endogenous genotoxic agents and processes as a basis of 83.spontaneous carcinogenesis. Mutât.Res., 218: 287-295, 1990.

57. Breimer, L. H. Molecular mechanisms of oxygen radical carcinogenesis andmutagenesis: the role of DNA base damage. Mol. Carcinog., 3: 188-197,1990.

58. Ames, B. N., and Gold, L. S. Too many rodent carcinogens: mitogenesis 84.increases mutagenesis. Science (Washington DC), 249: 970-971, 1990.

59. Floyd, R. A. The role of 8-hydroxyguanine in carcinogenesis. Carcinogenesis 85.(Lond.), //: 1447-1450, 1990.

60. Preston-Martin, S., Pike, M. C., Ross, R. K.. Jones, P. A., and Henderson,B. E. Increased cell division as a cause of human cancer. Cancer Res., 50:7415-7421, 1990. 86.

61. Cerutti, P. A, and Trump, B. F. Inflammation and oxidative stress incarcinogenesis. Cancer Cells (Cold Spring Harbor), 3: 1-7, 1991.

62. Barrett, J. C. The relationship between mutagenesis and carcinogenesis. In: 87.J. Brugge, T. Curren, E. Harlow, and F. McCormick (eds.). Origins ofHuman Cancer. Cold Spring Harbor, NY: Cold Spring Harbor LaboratoryPress, in press, 1991. 88.

63. Weinstein, I. B. Mitogenesis is only one factor in carcinogenesis. Science 89.(Washington DC), 251: 387-388, 1991.

64. Miller, J. A. Carcinogenesis by chemicals: an overview—G. H. A. ClowesMemorial Lecture. Cancer Res., 30: 559-576, 1970. 90.

65. Garner, R. C., Martin. C. N., and Clayson, D. B. Carcinogenic aromaticamines and related compounds in chemical carcinogens. In: A. Searle (ed.), 91.Chemical Carcinogens, pp. 175-276. Washington, D. C.: American Chemical Society, 1984.

66. Stowers, S. J., and Anderson. M. W. Formation and persistence ofbenzo(a)pyrene metabolite—DNA adducts. Environ. Health Perspect., 62: 92.31-39, 1985.

67. Stoner, G. D., Daniel, F. B., Schenck, K. M., Schul, H. A., Sandwisch, D.W., and Gohara, A. F. DNA binding and adduci formation of aflatoxin Bl 93.in cultured human and animal tracheobronchial and bladder tissues. Carcinogenesis (Lond.), 3: 1345-1348, 1982.

68. Williams, R. T. Inter-species variations in the metabolism of xenobiotics.Biochem. Soc. Trans., 2: 359-377, 1974. 94.

69. Berenblum, L, and Shubik, P. A new quantitative approach to the study ofthe stages of chemical carcinogenesis in the mouse skin. Br. J. Cancer, I:383-391, 1947. 95.

70. Gonzalez, F. J., Crespi, C. L., and Gelboin, H. V. DNA-expressed humancytochrome P450s: a new age of molecular toxicology and human risk 96.assessment. Mutât.Res.. 247: 113-127, 1991.

71. Huberman, E.. and Sachs, L. Metabolism of the carcinogenic hydrocarbonbenzo(a)pyrene in human fibroblast and epithelial cells. Int. J. Cancer, ¡I:412-418,1973. 97.

72. Belinsky. S. A., Dolan, M. E., White. C. M., Maronpot. R. R., Pegg, A. E.,

5037s

and Anderson, M. W. Cell specific differences in O6-methylguanine-DNAmethyltransferase activity and removal of CX'-methylguanine in rat pulmonary cells. Carcinogenesis (Lond.), 9: 2053-2058, 1988.Autrup, H., and Harris, C. C. Metabolism of chemical carcinogens byhuman tissues. In: C. C. Harris and H. Autrup (eds.). Human Carcinogenesis, pp. 169-194. New York: Academic Press, Inc., 1983.Conney, A. H., Welch, R.. Kmit/man. R., Chang, R., Jacobson, M., Munro-Faure, A. D., Peck, A. W., Bye, A., Poland, A., Poppers, P. J., Finster, M.,and Wolff, J. A. Effects of environmental chemicals on the metabolism ofdrugs, carcinogens, and normal body constituents in man. Ann. NY Acad.Sci., 179: 155-172, 1971.Harris, C. C. Interindividual variation among humans in carcinogen metabolism, DNA adduci formation and DNA repair. Carcinogenesis (Lond.), 10:1563-1566, 1989.Nebert, D. W., Nelson, D. R. Coon, M. J., Estabrook, R. W., Feyereisen,R., Fujii-Kuriyama, Y., Gonzalez, F. J., Guengerich, F. P., Gunsalus, I. C.,Johnson, E. F., Loper, J. C., Sato, R., Waterman, M. R., and Waxman, J.D. The P450 superfamily: update on new sequences, gene mapping andrecommended nomenclature. DNA Cell. Biol., 10: 1-20, 1991.Blum, M., Grant, D. M., McBride, W., Heim, M., and Meyer, U. A. Humanan lamine N-acetyltransferase genes: isolation, chromosomal localization,and functional expression. DNA Cell. Biol., 9: 193-203, 1990.Skoda, R. C.. Demierre, A., McBride, O. W., Gonzalez, F. J., and Meyer,U. A. Human microsomal xenobiotic epoxide hydrolase. ComplementaryDNA sequence, complementary DNA-directed expression in COS-1 cells,and chromosomal localization. J. Biol. Chem., 263: 1549-1554, 1988.Davies, R. L., Crespi. C. L., Rudo, K., Turner, T. R., and Langenbach, R.Development of a human cell line by selection and drug-metabolizing genetransfection with increased capacity to activate promutagens. Carcinogenesis(Lond.), 10: 885-891, 1989.Jaiswal, A. K., McBride, O. W., Adesnik, M., and Nebert, D. W. Humandioxin-inducible cytosolic NAD(P)H:menadione oxidoreductase. cDNA sequence and localization of gene to chromosome 16. J. Biol. Chem., 263:13572-13578.1988.Seidegard. J., Vorachek, W. R., Pero, R. W.. and Pearson, W. R. Hereditarydifferences in the expression of the human glutathione transferase active ontrans-stilbene oxide are due to a gene deletion. Proc. Nati. Acad. Sci. USA,«5:7293-7297, 1988.Gasser. R., Tynes, R. E., Lawton, M. P., Korsmeyer, K. K., Ziegler, D. M.,and Philpot, R. M. The flavin-containing monooxygenase expressed in pigliver: primary' sequence, distribution, and evidence for a single gene. Biochemistry1. 29:119-124, 1990.

Langenbach, R., Crespi, C., Davies, R., Rudo, K., Smith, P., Hansen, S.,Ross, J., Siegfried, J., and Nesnow, S. Transfection of cytochrome P450cDNAs into mammalian cells used in mutation and transformation assays.In: M. L. Mendelsohn and R. J. Albertini (eds.). Mutation and the Environment, Part D, pp. 239-248. New York: Wiley-Liss, 1990.Singer, B., and Grunberger, D. Molecular Biology of Mutagens and Carcinogens. New York: Plenum Publishing Corp., 1983.Preston, R. J. Mechanisms of induction of specific chromosomal alterations.In: B. M. Sutherland and A. D. Woodhead (eds.), DNA Damage and Repairin Human Tissues, pp. 329-336. New York: Plenum Publishing Corp.,1990.Lavi, S. Carcinogen-mediated amplification of viral DNA sequences insimian virus 40-transformed Chinese hamster embryo cells. Proc. Nati.Acad. Sci. USA, 78: 6144-6148, 1981.Brown, K.. Buchmann, A., and Balmain. A. Carcinogen-induced mutationsin the mouse c-Ha-raj gene provide evidence of multiple pathways for tumorprogression. Proc. Nati. Acad. Sci. USA, 87: 538-542, 1990.Barbacid, M. raj genes. Annu. Rev. Biochem., 56: 779-827, 1987.Newcomb, E. W., Diamond, L. E., Sloan, S. R., Corominas, M., Guerrerro,I., and Pellicer, A. Radiation and chemical activation of ras oncogenes indifferent mouse strains. Environ. Health Perspect., 81: 33-37, 1989.Balmain, A., and Brown, K. Oncogene activation in chemical carcinogenesis.Adv. Cancer Res., 51: 147-182. 1988.Yamasaki. H., Hollstein, M., Martel, N., Cabrai, J. R., Calendo, D., andTomatis, L. Transplacental induction of a specific mutation in fetal Ha-rajand its critical role in post-natal carcinogenesis. Int. J. Cancer, 40: 818-822, 1987.Sukumar, S. An experimental analysis of cancer: role of ras oncogenes inmultistep carcinogenesis. Cancer Cells (Cold Spring Harbor), 2: 199-204,1990.Bailleul, B., Surani, M. A.. White, S., Barton, S. C., Brown, K., Blessing,M., Jorcano, J., and Balmain, A. Skin hyperkeratosis and papilloma formation in transgenic mice expressing a ras oncogene from a suprabasalkeratin promoter. Cell, 62: 697-708, 1990.Leder, A.. Kuo, A., Cardiff, R. D., Sinn, E., and Leder, P. v-Ha-ras transgeneabrogates the initiation step in mouse skin tumorigensis: effects of phorbolesters and retinoic acid. Proc. Nati. Acad. Sci. USA, 87: 9178-9182, 1990.KHudson. A. G., Jr. Mutation and cancer: statistical study of retinoblastoma.Proc. Nati. Acad. Sci. USA, 68: 820-823, 1971.Cavenee, W. K., Dryja, T. P., Phillips, R. A., Benedict, W. F., Godbout,R., Gallic. B. L., Murphree, A. L., Strong, L. C., and White, R. L.Expression of recessive alíelesby chromosomal mechanisms in retinoblastoma. Nature (Lond.), 305: 779-784, 1983.Sapienza, C. Genome imprinting, cellular mosaicism and carcinogenesis.Mol. Carcinog., 3: 118-121, 1990.

on April 2, 2020. © 1991 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Page 16: Chemical and Physical Carcinogenesis: Advances …...CARCINOGENESIS Table 1 Examples of advances in chemical and physical carcinogenesis" A. Cancer etiology I. Life-style factors (tobacco

CARCINOGENESIS

98. Scheffner. M.. Wcrness. B. A.. Huibregtse. J. M., Levine. A. J.. and Howley.P. M. The E6 oncoprotein encoded by human papillomavirus types 16 and18 promotes the degradation of p53. Cell. 63: 1129-1136, 1990. 125.

99. Herskowitz, I. Functional inactivation of genes by dominant negative mutations. Nature (Lond.), 329: 219-222, 1987. 126.

100. Green, M. R. When the products of oncogenes and anti-oncogenes meet.Cell, 56: 1-3, 1989. 127.

101. Waldren, C, Correli, L., Sognier, M. A., and Puck, T. T. Measurement oflow levels of x-ray mutagenesis in relation to human disease [publishederratum appears in Proc. Nati. Acad. Sci. USA 84 (10):3364. 1987. Proc.Nail. Acad. Sci. USA, 83: 4839-4843, 1986.

102. Gray. J. W., Lucas, L., Kallioniemi. O.. Kallioniemi, A.. Kuo. W. L., Pinkel, 128.D.. Tkachuk. D., and Weier, U. Applications of fluorescence in situ hybridization in biological dosimetry. In: B. L. Gledhill and F. Mauro (eds.).Trends in Biological Dosimetry. New York: Wiley-Liss. in press, 1991.

103. Wolff. S. Biological dosimetry with cytogenetic endpoints. In: B. L. Gledhill 129.and F. Mauro (eds.). Trends in Biological Dosimetry. New York: Wiley-Liss, in press, 1991.

104. Loeb, L. A., and Preston, B. D. Mutagenesis by apurinic/apyrimidinic sites. 130.Annu. Rev. Genet., 20: 201-230. 1986.

105. Basu, A. K.. and Essigmann, J. M. Site specifically modified oligodeoxy-nucleotides as probes for the structural and biological effects of DNA- 131.damaging agents. Chem. Res. Toxicol., /: 1-18, 1988.

106. Skandalis, A., and Glickman. B. W. Endogenous gene systems for the studyof mutational specificity in mammalian cells. Cancer Cells (Cold Spring 132.Harbor), 2: 79-83, 1990.

107. Sarasin, A. Use of shuttle vectors for the analysis of DNA damage processing 133.in mammalian cells. In: Mutation Research. Vol. 220, Nos. 2/3. Amsterdam:Elsevier Science Publishers BV. 1989.

108. Sarasin, A. Shuttle vectors for studying mutagenesis in mammalian cells. J.Photochem. Pholobiol. B Biol., 3: 143-155, 1989. 134.

109. Mäher.V. M., Yang. J. L.. Mah, M. C., and McCormick, J. J. Comparingthe frequency and spectra of mutations induced when an SV-40 based shuttlevector containing covalenti) bound residues of structurally-related carcinogens replicates in human cells. Mutât.Res., 220: 83-92, 1989.

110. Basu, A. K., and Essigmann, J. M. Site-specifically alkylated oligodeoxy- 135.nucleotides: probes for mutagenesis, DNA repair and the structural effectsof DNA damage. Mutât.Res., 233: 189-201, 1990. 136.

111. Mazur. M.. Gordon, A. J. F., Bernelot-Moens, C., and Glickman, B. W.Comparison of the mutational specificities exhibited by BPDE in Esche-richia coli and CHO cells. In: M. W. Lambert and J. Laval (eds.), DNARepair Mechanisms and their Biological Implications in Mammalian Cells,New York: Plenum Publishing Corp.. 1989. 137.

112. Yang, J. L.. Chen, R. H., Mäher,V. M., and McCormick, J. J. Kinds andlocation of mutations induced by (±)-7tf,8«-dihydroxy-9n,lOn-epoxy-7,8,9,10-tetrahydrobenzo[a]pyrene in the coding region of the hypoxanthine 138.(guanine) phosphoribosyltransferase gene in diploid human fibroblasts. Car-cinogenesis (Lond.), 12: 71-75, 1991.

113. Carothers, A. M., Urlaub, G., Mucha, J.. Harvey, R. G.. Chasin, L. A., and 139.Grunberger, D. Splicing mutations in the CHO DHFR gene preferentially-induced by (±)-3«,4/}-dihydroxy-1n,2n-epoxy-1,2.3,4-tetrahydrobenzo[c]phenanthrene. Proc. Nati. Acad. Sci. USA, «7:5464-5468, 1990. 140.

114. Alberimi, R. J., Sullivan, L. M., Berman. J. K.. Greene, C. J., Stewart, J.A., Silveira, J. M., and O'Neill, J. P. Mutagenicity monitoring in humans 141.

by autoradiographic assay for mutant T lymphocytes. Mutât.Res., 204:481-492, 1988.

115. Loveless, A. Possible relevance of O-6 alkylation of deoxyguanosine to themutagenicity and carcinogenicity of nitrosamines and nitrosamides. Nature(Lond.), 225:206-207, 1969. ' 142.

116. Loechler, E. L., Green, C. L., and Essigmann, J. M. In vivo mutagenesis byO'-methylguanine built into a unique site in a viral genome. Proc. Nail.Acad. Sci. USA, 81: 6271-6275, 1984. 143.

117. Zarbl. H.. Sukumar, S., Arthur, A. V., Martin-Zanca, D., and Barbacid, M.Direct mutagenesis of Ha-rai-1 oncogenes by /V-nitroso-A'-methylurea during initiation of mammary carcinogenesis in rats. Nature (Lond.), 315: 382- 144.385, 1985.

118. Doniger, J., Notario. V., and DiPaolo, J. A. Carcinogens with diversemutagenic activities initiate neoplastic guinea pig cells that acquire the same 145.N-ras point mutation. J. Biol. Chem., 262: 3813-3819, 1987.

119. Pelling. J. C.. Neades, R., and Strawhecker. J. Epidermal papillomas andcarcinomas induced in uninitiated mouse skin by tumor promoters alonecontain a point mutation in the 6Ist codon of the Ha-ras oncogene. Carci- 146.nogenesis (Lond.). 9: 665-667. 1988.

120. Brookes, P. Chemical carcinogens and raj gene activation. Mol. Carcinog.,2:305-307, 1989.

121. Topal, M. D. DNA repair, oncogenes and carcinogenesis. Carcinogenesis 147.(Lond.), 9:691-696, 1988.

122. Weinstein, I. B., Jeffrey, A. M., Jennette, K. W., Blobstein, S. H., Harvey,R. G., Harris, C. C., Autrup, H., Kasai, H., and Nakanishi, K.Benzo(a)pyrene diol epoxides as intermediates in nucleic acid binding in 148.vitro and in vivo. Science (Washington DC), 193: 592-595, 1976.

123. Dipple. A.. Pigott. M., Moschel, R. C., and Costantino. N. Evidence thatbinding of 7,12-dimethylbenz(a)anthracene to DNA in mouse embryo cellcultures results in extensive substitution of both adenine and guanine resi- 149.dues. Cancer Res., «.-4132-4135, 1983.

124. Wood, M. L.. Dizdaroglu, M., Gajewski, E.. and Essigmann, J. M. Mechanistic studies of ionizing radiation and oxidative mutagenesis: genetic

5038s

effects of a single 8-hydroxyguanine (7-hydro-8-oxoguanine) residue insertedat a unique site in a viral genome. Biochemistry, 349: 7024-7032, 1990.I iiiilalil. T., and Nyberg. B. Rate of depurination of native deoxyribonucleicacid. Biochemistry. //: 3610-3618. 1972.Soussi. T., Carón de Fromentel, C., and May, P. Structural aspects of thep53 protein in relation to gene evolution. Oncogene. 5: 945-952, 1990.Nigro, J. M., Baker, S. J., Preisinger, A. C., Jessup, J. M., Hosteller, R..deary, K., Bigner, S. H., Davidson. N., Baylin, S., Devilee, P., Glover, T.,Collins, F. S., Weston, A.. Modali. R., Harris, C. C., and Vogelstein, B.Mutations in the p53 gene occur in diverse human tumor types. Nature(Lond.), 342: 705-708, 1989.Takahashi, T., Ñau. M. M.. Chiba. L, Birrer, M. J., Rosenberg. R. K..Vinocour, M., Leviti, M., Pass, H., Gazdar, A. F., and Minna, J. D. p53: afrequent target for genetic abnormalities in lung cancer. Science (Washington DC). 246: 491-494. 1989.Iggo. R., Gatter. K., Bartek. J., Lane. D.. and Harris. A. L. Increasedexpression of mutant forms of pS3 oncogene in primary lung cancer. Lancet.335: 675-679, 1990.Mulligan, L. M., Matlashewski. G.. Scrable. H. J., and Cavenee, W. K.Mechanisms of p53 loss in human sarcomas. Proc. Nati. Acad. Sci. USA,«7:5863-5867, 1990.Hollstein. M.. Metcalf. R. A., Welsh. J.. Montesano. R.. and Harris, C. C.Frequent mutation of the p53 gene in human esophagcal cancer. Proc. Nati.Acad. Sci. USA, 87: 9958-9961, 1990.Hollstein, M.. Sidransky. D.. Vogelstein. B., and Harris, C. C. p53 mutations in human cancers. Science (Washington DC), 253: 49-53, 1991.Rodenhuis, S., Van de Wetering, M. L., Mooi, W. J., Evers, S. G., vanZandwijk, N., and Bos, J. L. Mulational activation of the K-ras oncogene.A possible pathogenetic factor in adenocarcinoma of the lung. N. Engl. J.Med.,317: 929-935, 1987.Slebos, R. J., Kibbelaar. R. E., Dalesio. O., Kooistra. A., Stam, J., Meijer,C. J., Wagenaar, S. S., Vanderschueren. R. G.. van Zandwijk. N.. Mooi,W. J., Bos. J. L.. and Rodenhuis. S. K-ros oncogene activation as aprognostic marker in adenocarcinoma of the lung. N. Engl. J. Med.. 323:561-565, 1990.Bos. J. L. ras oncogenes in human cancer: a review. Cancer Res., 49: 4682-4689, 1989.Chiba. !.. Takahashi, T., Ñau, M. M., D'Amico, D., Curici. D. T., Mitsu-domi. T.. Buchhagen, D. L.. Carbone, D., Piantadosi, S., Koga, H., Reiss-man, P. T.. Slamon. D. J.. Holmes. E. C.. and Minna. J. D. Mutations inthe p53 gene are frequent in primary, resected non-small cell lung cancer.Oncogene. 5: 1603-1610, 1990.Hsu, 1. C., Metcalf. R. A., Sun, T., Welsh, J., Wang, N. J., and Harris, C.C. p53 gene mulational hotspot in human hepatocellular carcinomas fromQidong, China. Nature (Lond.), 350:427-428. 1991.Bressac, B., Kew, M., Wands, J., and Ozturk. M. Selective G to T mutationsof p53 gene in hepatocellular carcinoma from southern Africa. Nature(Lond.), 550:429-431, 1991.McMahon, G., Davis. E. F., Huber, L. J., Kim, Y., and Wogan, G. N.Characterization of c-Ki-ras and N-ras oncogenes in aflaloxin Bi-inducedrat liver tumors. Proc. Nati. Acad. Sci. USA. 87: 1104-1108, 1990.Wainscoat. J. S., and Fey, M. F. Assessment of clonali!) in human tumors:a review. Cancer Res., 50: 1355-1360, 1990.Malkin. D., Li, F. P., Strong. L. C.. Fraumeni. J. F., Nelson. C. E., Kim,D. H.. Kassel, J.. Gryka, M. A., Bischoff, F. Z.. Tainsky, M. A., and Friend,S. H. Germ line p53 mutations in a familial syndrome of breast cancer,sarcomas, and other neoplasms. Science (Washington DC), 250: 1233-1238, 1990.Srivastava, S., Zou, Z. Q., Pirollo, K.. Blattner, W. A., and Chang, E. H.Germline transmission of a mutated p53 in a cancer-prone family with Li-Fraumeni syndrome. Nature (Lond.), 348: 747-749, 1990.Baker, S. J., Markowitz, S., Fearon. E. R., Willson, J. K.. and Vogelstein,B. Suppression of human colorectal carcinoma cell growth by wild-type p53.Science (Washington DC). 249:912-915, 1990.Chen, P. L., Chen, Y., Bookstein, R., and Lee, W. H. Genetic mechanismsof tumor suppression by the human p53 gene. Science (Washington DC),250: 1576-1580, 1991.Mercer, W. E.. Shields, M. T., Amin, M., Sauve, G. J., Appella, E.. Romano,J. W., and Ullrich, S. J. Negative growth regulation in a glioblastoma tumorcell line that conditionally expresses human wild-type p53. Proc. Nail. Acad.Sci. USA, 87: 6166-6170. 1990.Diller, L., Kassel, J., Nelson, C. E., Gryka, M. A., Litwak, G., Gebhardt.M., Bressac, B., Ozturk, M.. Baker, S. J., Vogelslein, B., and Friend, S. H.p53 functions as a cell cycle control protein in osteosarcomas. Mol. Cell.Biol.. 10: 5772-5781, 1990.Huang, H. J., Yee, J. K., Shew, J. Y., Chen, P. L., Bookstein, R., Friedmann,T., Lee, E. Y., and Lee, W. H. Suppression of the neoplastic phenotype byreplacement of the RB gene in human cancer cells. Science (WashingtonDC), 242: 1563-1566. 1988.Sumegi, J., Uzvolgyi, E.. and Klein. G. Expression of the Rb gene underthe control of MuLV-LTR suppresses tumorigenicity of WERl-Rb-27 ret-inoblastoma cells in immunodefective mice. Cell Growth Differentiation, /:247-250, 1990.Mercer, W. E., Shields, M. T.. Lin, D., Appella. E., and Ullrich, S. J.Growth suppression induced by wild type p53 protein is accompanied byselective down regulation of proliferating cell nuclear antigen (PCNA)expression. Proc. Nati. Acad. Sci. USA, in press, 1991.

on April 2, 2020. © 1991 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Page 17: Chemical and Physical Carcinogenesis: Advances …...CARCINOGENESIS Table 1 Examples of advances in chemical and physical carcinogenesis" A. Cancer etiology I. Life-style factors (tobacco

CARCINOGENESIS

150. Bailleul. B., Brown, K., Ramsden, M., Akhurst, R. J., Fee, F., and Balmain, 177.A. Chemical induction of oncogene mutations and growth factor activity inmouse skin carcinogenesis. Environ. Health Perspect.. */: 23-27, 1989. 178.

151. Kumar. R.. Sukumar. S.. and Barbacid, M. Activation of raj oncogenespreceding the onset of neoplasia. Science (Washington DC), 248: 1101-1104, 1990.

152. Kumar. R.. Medina, D., and Sukumar. S. Activation of H-ras oncogenes in 179.preneoplastic mouse mammary tissues. Oncogene, 5: 1271-1277. 1990.

153. Sinn, E., Muller, W., Pattengale, P., Tepler, I., Wallace. R.. and Leder. P.Coexpression of MMTV/v-Ha-ras and MMTV/c-m>'c genes in transgenic 180.mice: synergistic action of oncogenes in vim. Cell, 49: 465-475, 1987.

154. Yamasaki, H. Gap junctional intercellular communication and carcinogenesis. Carcinogenesis (Lond.). //: 1051-1058. 1990.

155. Missero, C.. Filvaroff, E., and Dotto, G. P. Induction of transforminggrowth factor beta 1 resistance by the EIA oncogene requires binding to a 181.specific set of cellular proteins. Proc. Nati. Acad. Sci. USA. SS: 3489-3493,1991.

156. Amstad, P.. Reddel. R. R.. Pfeifer. A.. Malan-Shibley, L.. Mark, G. E.. and 182.Harris. C. C. Neoplastic transformation of a human bronchial epithelial cellline by a recombinant retrovirus encoding viral harvey ras. Mol. Carcinog., 183./: 151-160, 1988.

157. Reddel, R. R., Ke, Y., Kaighn, M. E., Malan-Shibley, L.. Lechner, J. F.,Rhim, J. S., and Harris, C. C. Human bronchial epithelial cells neoplasticallytransformed by v-Ki-ras: altered response to inducers of terminal squamous 184.differentiation. Oncogene Res.. 3: 401-408, 1988.

158. Pfeifer. A.. Mark, G. E., Malan-Shibley. L., Graziano. S. L.. Amstad. P..and Harris, C. C. Cooperation of c-rai-1 and c-myc protooncogenes in theneoplastic transformation of SV40 T-antigen immortalized human bron- 185.chial epithelial cells. Proc. Nati. Acad. Sci. USA, 86: 10075-10079, 1989.

159. Hynes, R. O., and Plantefaber, L. C. Integrin receptors for extracellularmatrix and their involvement in oncogenic transformation. In: J. Brugge, 186.T. Curren, E. Harlow, and F. McCormick (eds.). Origins of Human Cancer,New York: Cold Spring Harbor Laboratory Press, in press, 1991.

160. Sporn, M. B., and Todaro, G. J. Autocrine secretion and malignant trans- 187.formation of cells. N. Engl. J. Med., 303: 878-880, 1980.

161. Pardee. A. B.. Medrano, E. E., and Rossow, P. W. In: M. Ritzen, A. Aperia, 188.K. Hall, A. Larsson. A. Zetterberg. and R. Zetterstrom (eds.). The Biologyof Normal Human Growth, pp. 59-70. New York: Raven Press, Ltd.. 1981. 189.

162. Parchment, R. E., and Pierce, G. B. Polyamine oxidation, programmed celldeath, and regulation of melanoma in the murine embryonic limb. CancerRes., 49: 6680-6686. 1989.

163. McDonnell, T. J.. and Korsmeyer, S. J. Progression from lymphoid hyper- 190.plasia to high-grade malignant lymphoma in mice transgenic for the t(14;18).Nature (Lond.), 349: 254-256, 1991.

164. Gregory, C. D., Dive, C., Henderson. S.. Smith, C. A., Williams, G. T., 191.Gordon, J., and Rickinson, A. B. Activation of Epstein-Barr virus latentgenes protects human B cells from death by apoptosis. Nature (Lond.), 349: 192.612-614, 1991.

165. Giancotti, F. G., and Ruoslahti, E. Elevated levels of the «5fi,fibronectin 193.receptor suppress the transformed phenotype of Chinese hamster ovarycells. Cell. 60: 849-859, 1990.

166. Cuttitta, F., Carney, D. N., Mulshine, J., Moody, T. W., Fedorko, J.,Fischler, A., and Minna. J. D. Bombesin-like peptides can function as 194.autocrine growth factors in human small-cell lung cancer. Nature (Lond.),316: 823-826, 1985.

167. Mendelsohn, J. Growth factor receptors as targets for antitumor therapy 195.with monoclonal antibodies. Prog. Allergy, 45: 147-160, 1988.

168. Kasid, U., Pfeifer, A., Brennan, T., Beckett, M., Weichselbaum, R. R.,Dritschilo, A., and Mark, G. E. Effect of antisense c-ra/-l on tumorigenicity 196.and radiation sensitivity of a human squamous carcinoma. Science (Washington DC), 243: 1354-1356, 1989.

169. Rivera. R. T.. Pasion, S. G., Wong, D. T., Fei, Y. B., and Biswas, D. K. 197.Loss of tumorigenic potential by human lung tumor cells in the presence ofantisense RNA specific to the ectopically synthesized a subunit of humanchorionic gonadotropin. J. Cell Biol., 108: 2423-2434, 1989. 198.

170. McManaway, M. E., Neckers, L. M., Loke, S. L., al-Nasser, A. A., Redner,R. L., Shiramizu, B. T., Goldschmidts, W. L., Huber, B. E., Bhatia, K., andMagrath, I. T. Tumour-specific inhibition of lymphoma growth by anantisense oligodeoxynucleotide. Lancet, 335: 808-811,1990. 199.

171. Reed, J. C.. Cuddy. M., Haldar, S., Croce, C, Nowell, P., Makover, D.,and Bradley, K. BCL2-mediated tumorigenicity of a human T-lymphoid cell 200.line: synergy with M YC and inhibition by BCL2 antisense. Proc. Nati. Acad.Sci. USA, 87: 3660-3664. 1990. 201.

172. Tidd, D. M. A potential role for antisense oligonucleotide analogues in thedevelopment of oncogene targeted cancer chemotherapy. Anticancer Res.,10: 1169-1182, 1990. 202.

173. Cohen, P. The structure and regulation of protein phosphatases. Annu. Rev.Biochem., 58: 453-508, 1989.

174. Farago, A., and Nishizuka, Y. Protein kinase C in transmembrane signal)- 203.ing. FEBS Lett.. 268: 350-354. 1990.

175. Angel. P., Imagawa, M., Chiù,R., Stein. B., Imbra, R. J.. Rahmsdorf, H.J., Jonat, C., Herrlich, P., and Karin, M. Phorbol ester-inducible genes 204.contain a common as element recognized by a TPA-modulated trans-actingfactor. Cell, 49: 729-739, 1987.

176. Weinstein. I. B. The origins of human cancer: molecular mechanisms ofcarcinogenesis and their implications for cancer prevention and treatment— 205.Twenty-seventh G. H. A. Clowes Memorial Award Lecture. Cancer Res.,48: 4135-4143, 1988.

5039s

Fujiki, H., and Suganuma, M. Significance of new environmental tumorpromoters. J. Environ. Sci. Health, C7: 1-51, 1989.Haystead, T. A., Sim, A. T., Carling, D., Honnor, R. C., Tsukitani, Y.,Cohen, P., and Hardie, D. G. Effects of the tumour promoter okadaic acidon intracellular protein phosphorylation and metabolism. Nature (Lond.),i J 7: 78-81, 1989.Takai. A.. Bialojan, C., Troschka, M., and Ruegg, J. C. Smooth musclemyosin phosphatase inhibition and force enhancement by black spongetoxin. FEBS Lett., 217: 81-84, 1987.Cole, J., Waugh, A. P. W., Beare, D. M., Sala-Trepat, M., Stephens, G.,and Green, M. H. L. HPRT mutant frequencies in circulating lymphocytes:population studies using normal donors, exposed groups and cancer-pronesyndromes. In: B. L. Gledhill and F. Mauro (eds.), Trends in BiologicalDosimetry. New York: Wiley-Liss, in press, 1991.Menichini, P., and Abbondandolo. A. Somatic gene mutations in humans.In: B. L. Gledhill and F. Mauro (eds.). Trends in Biological Dosimetry. NewYork: Wiley-Liss. in press, 1991.Ehrlich, M., and Wang, R. Y. 5-Methylcytosine in eukaryotic DNA. Science(Washington DC), 212: 1350-1357, 1981.Ehrlich, M., Zhang, X. Y., and Inamdar, N. M. Spontaneous deaminationof cytosine and 5-methylcytosine residues in DNA and replacement of 5-methylcytosine residues with cytosine residues. Mutât.Res., 238: 277-286,1990.Phear, G., Nalbantoglu, J., and Meuth, M. Next-nucleotide effects inmutations driven by DNA precursor pool imbalances at the aprt locus ofChinese hamster ovary cells. Proc. Nati. Acad. Sci. USA, 84: 4450-4454,1987.Liu, P. K., Chang, C. C, Trosko, J. E., Dube, D. K.. Martin, G. M., andLoeb, L. A. Mammalian mutator mutant with an aphidicolin-resistant DNApolymerase a. Proc. Nati. Acad. Sci. USA, 80: 797-801, 1983.Kunz, B. A., Kang. X., and Kohalmi, L. The yeast radlS mutator specificallyincreases G:C—>T:Atransversions without reducing correction of G-A or C-T mismatches to G:C pairs. Mol. Cell. Biol., //: 218-225, 1991.Loeb, L. A., Springgate, C. F., and Battula, N. Errors in DNA replicationas a basis of malignant changes. Cancer Res., 34: 2311-2321, 1974.Chorazy, M. Sequence rearrangements and genome instability. A possiblestep in carcinogenesis. J. Cancer Res. Clin. Oncol., 109: 159-172, 1985.German, J. Bloom's syndrome. X. The cancer proneness points to chro

mosome mutation as a crucial event in human neoplasia. In: J. German(ed.), Chromosome Mutation and Neoplasia, pp. 347-357. New York: AlanR. Liss, Inc., 1983.Kern, S. E., Fearon, E. R., Tersmette, K. W., Enterline, J. P., Leppert, M.,Nakamura, Y., White, R., Vogelstein, B., and Hamilton, S. R. Allelic lossin colorectal carcinoma. JAMA, 261: 3099-3103, 1989.Nowell, P. C. Mechanisms of tumor progression. Cancer Res., 46: 2203-2207, 1986.Wolman, S. R. Karyotypic progression in human tumors. Cancer MetastasisRev., 2: 257-293, 1983.Barrett, J. C., Tsutsui, T., Tlsty, T., and Oshimura, M. Role of geneticinstability in carcinogenesis. In: C. C. Harris and L. A. Liotta (eds.). GeneticMechanisms in Carcinogenesis and Tumor Progression, pp. 97-114. NewYork: Wiley-Liss, 1990.Seshadri, R., Kutlaca. R. J., Trainor, K., Matthews, C., and Morley, A. A.Mutation rate of normal and malignant human lymphocytes. Cancer Res.,47:407-409, 1987.Elmore, E., Kakunaga, T., and Barrett, J. C. Comparison of spontaneousmutation rates of normal and chemically transformed human skin fibro-blasts. Cancer Res., 43: 1650-1655, 1983.Cifone, M. A., and Fidler, I. J. Increasing metastatic potential is associatedwith increasing genetic instability of clones isolated from murine neoplasms.Proc. Nati. Acad. Sci. USA, 78: 6949-6952, 1981.Kendal, W. S., and Frost, P. Metastatic potential and spontaneous mutationrates: studies with two murine cell lines and their recently induced metastaticvariants. Cancer Res., 46: 6131-6135, 1986.Frost, P., Kendal, W., Hunt, B., and Ellis, M. The prevalence of ouabain-resistant variants after mutagen treatment. Lack of correlation between thefrequency of variant expression and the metastatic phenotype. Invasion &Metastasis, 8: 73-86, 1988.Sager, R. Mutation rates and mutational spectra in tumorigenic cell lines.Cancer Surv., 7:325-333, 1988.Goldberg, S., and Defendi, V. Increased mutation rates in doubly viraltransformed Chinese hamster cells. Somatic Cell. Genet., 5: 887-895,1979.Yamashina, K., and Heppner, G. H. Correlation of frequency of inducedmutation and metastatic potential in tumor cell lines from a single mousemammary tumor. Cancer Res., 45: 4015-4019, 1985.Chambers, A. F., Harris, J. G., and Grundy, J. S. Rates of generation ofMTX-resistant variants in cells temperature sensitive for malignant transformation. Somatic Cell Mol. Genet., 14: 253-259, 1988.Sager, R., Cadi, I. K., Stephens, L., and Grabowy, C. T. Gene amplification:an example of accelerated evolution in tumorigenic cells. Proc. Nati. Acad.Sci. USA, 82: 7015-7019. 1985.Tlsty, T. D., Margolin. B. H.. and Lum, K. Differences in the rates of geneamplification in nontumorigenic and tumorigenic cell lines as measured byLuria-Delbruck fluctuation analysis. Proc. Nati. Acad. Sci. USA, 86: 9441-9445, 1989.Tlsty, T. D. Normal diploid human and rodent cells lack a detectablefrequency of gene amplification. Proc. Nati. Acad. Sci. USA, 87: 3132-3136, 1990.

on April 2, 2020. © 1991 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Page 18: Chemical and Physical Carcinogenesis: Advances …...CARCINOGENESIS Table 1 Examples of advances in chemical and physical carcinogenesis" A. Cancer etiology I. Life-style factors (tobacco

CARCINOGENESIS

206. Wright. J. A., Smith. H. S.. Watt. F. M., Hancock, M. C.. Hudson, D. L., 233.and Stark. G. R. DNA amplification is rare in normal human cells. Proc.Nail. Acad. Sci. USA. 87: 1791-1795. 1990. 234.

207. Croce. C. M. Molecular biology of leukemias and lymphomas. ln:J. Brugge.T. Curren, E. Harlow, and F. McCormick (eds.). Origins of Human Cancer. 235.Cold Spring Harbor, NY: Cold Spring Harbor Laboratory press, in press,1991.

208. Hastie. N. D.. Dempster, M., Dunlop. M. G., Thompson, A. M., Green, D. 236.K., and Allshire, R. C. Telomere reduction in human colorectal carcinomaand with ageing. Nature (Lond.), 346: 866-868, 1990.

209. Schmid. M., Haaf. T., and Grunert. D. 5-Azacytidine-induced undercon- 237.densations in human chromosomes. Hum. Genet., 67: 257-263, 1984.

210. Apian. P. D.. Lombardi. D. P.. Ginsberg. A. M.. Cossman. J., Bertness, V.L., and Kirsch. I. R. Disruption of the human SCL locus by "illegitimate"V-(D)-J recombinase activity. Science (Washingtion DC). 250: 1426-1429. 238.1990.

211. Lundblad, V., and Szostak, J. W. A mutant with a defect in telomereelongation leads to senescence in yeast. Cell, 57: 633-643, 1989. 239.

212. McClintock, B. Chromosome organization and genie expression. ColdSpring Harbor Symp. Quant. Biol.. 16: 13-49, 1951.

213. Hartwell. L. H., and Weinert. T. A. Genetic control of mitotic fidelity inyeast and its relation to cancer. In: J. Brugge. T. Curren. E. Harlow. and F. 240.McCormick (eds.). Origins of Human Cancer. Cold Spring Harbor, NY:Cold Spring Harbor Laboratory Press, in press, 1991.

214. Bischoff. J. R., and Beach, D. Model systems for the study of p53 function.In: J. Brugge, T. Curren, E. Harlow, and F. McCormick (eds.), Origins of 241.Human Cancer. Cold Spring Harbor, NY: Cold Spring Harbor LaboratoryPress, in press, 1991.

215. Weinert, T. A., and Hartwell, L. H. Characterization of RAD9 of Saccha-romyces cererisiae and evidence that its function acts posttranslationally in 242.cell cycle arrest after DNA damage. Mol. Cell Biol., 10:6554-6564, 1990.

216. Uemura. T.. and Yanagida. M. Isolation of type I and II DNA topoisomerasemutants from fission yeast: single and double mutants show different phe- 243.notypes in cell growth and chromatin organization. EM BO J., 3: 1737-1744, 1984.

217. Uemura. T.. Ohkura. H., Adachi, Y., Morino, K., Shiozaki. K., and Yanagida, M. DNA topoisomerase II is required for condensation and separation 244.of mitotic chromosomes in S. pombe. Cell, 50:917-925, 1987.

218. Bischoff. F. Z.. Yim. S. O., Pathak. S.. Grant. G.. Siciliano, M. J., Giova-nella, B. C., Strong, L. C.. and Tainsky. M. A. Spontaneous abnormalitiesin normal fibroblasts from patients with Li-Fraumeni cancer syndrome: 245.aneuploidy and immortalization. Cancer Res., 50: 7979-7984, 1990.

219. Higginson, J.. and Muir, C. S. Environmental carcinogenesis: misconceptions and limitations to cancer control. J. Nati. Cancer Inst. (Bethesda). 63: 246.1291-1298, 1979.

220. Wynder, E. L.. and Reddy, B. S. Metabolic epidemiology of colorectalcancer. Cancer (Phila.), 34: 801-806. 1974.

221. Harris. C. C, Vahakangas. K., Autrup. H.. Trivers, G. E., Shamsuddin. A. 247.K. M., Trump. B. F., Boman. B. M.. and Mann. D. L. Biochemical andmolecular epidemiology of human cancer risk. In: D. Scarpelli, J. Craighead.and N. Kaufman (eds.). The Pathologist and the Environment, pp. 140- 248.167. Baltimore: Williams and Wilkins. 1985.

222. Perera, F. P., and Weinstein, I. B. Molecular epidemiology and carcinogen-DNA adduci detection: new approaches to studies of human cancer causa- 249.lion. J. Chronic. Dis., 35: 581-600, 1982.

223. Harris, C. C. Molecular epidemiology of human cancer in the 1990's. In:

B. L. Gledhill and F. Mauro (eds.). Trends in Biological Dosimetry. New 250.York: Wiley-Liss, in press, 1991.

224. Wogan, G. N. Markers of exposure to carcinogens. Environ. Health Perspect., «/:9-17, 1989. 251.

225. Alberimi, R. J., and Robison, S. H. Human population monitoring. In: A.P. Li and R. H. Heflich (eds.). Genetic Toxicology: A Treatise, pp. 373-418. Caldwell, NJ: Telford Press, 1991. 252.

226. Harris, C. C., Weston, A., Willey, J. C., Trivers, G. E., and Mann, D. L.Biochemical and molecular epidemiology of human cancer: indicators ofcarcinogen exposure, DNA damage, and genetic predisposition. Environ.Health Perspect.. 75: 109-119. 1987.

227. Sanders. B. M., Jay, M.. Draper. G. J., and Roberts, E. M. Non-ocular 253.cancer in relatives of retinoblastoma patients. Br. J. Cancer, 60: 358-365,1989.

228. Cleaver. J. E. Do we know the cause of xeroderma pigmentosum. Carcino- 254.genesis (Lond.). / 7:875-882. 1990.

229. Nicotera, T. M., Notaro, J.. Notaro, S., Schumer, J.. and Sandberg, A. A.Elevated Superoxide dismutase in Bloom's syndrome: a genetic condition ofoxidalive stress. Cancer Res., 49: 5239-5243, 1989.

230. Carlwright, R. A.. Glashan. R. W.. Rogers, H. J., Ahmad, R. A.. Barham- 255.Hall. D., Higgins. E.. and Kahn. M. A. Role of iV-acetyltransferase pheno-types in bladder carcinogenesis: a pharmacogenetic epidemiological ap- 256.proach lo bladder cancer. Lancet. 2: 842-845. 1982.

231. Ayesh. R., Idle. J. R., Ritchie, J. C.. Crothers, M. J., and Hetzel, M. R. 257.Metabolic oxidation phenotypes as markers for susceptibility to lung cancer.Nature (Lond.), 312: 169-170. 1984.

232. Caporaso, N. E., Tucker. M. A.. Hoover, R., Hayes, R. B., Pickle, L. W., 258.Issaq, H.. Muschik, G., Green-Gallo. L., Buivys. D.. Aisner. S., Resau. J..Trump, B. F., Tollerud. D., Weston, A., and Harris, C. C. Lung cancer andthe debrisoquine metabolic phenotype. J. Nail. Cancer Inst. (Bethesda), 85: 259.1264-1272, 1990.

5040s

Hanawalt, P. C. Preferential DNA repair in expressed genes. Environ.Health Perspect.. 76:9-14. 1987.Dolan. M. E.. Oplinger. M.. and Pegg. A. E. Sequence specificity of guaninealkylation and repair. Carcinogenesis (Lond.), 9: 2139-2143. 1988.Mattes. W. B.. Hartley, J. A.. Kohn, K. W., and Matheson, D. W. GR-richregions in genomes as targets for DNA alkylation. Carcinogenesis (Lond.),9:2065-2072, 1988.Conney, A. H. Induction of microsomal enzymes by foreign chemicals andcarcinogenesis by polycyclic aromatic hydrocarbons: G. H. A Clowes Memorial Lecture. Cancer Res., ¥2:4875-4917. 1982.Fujino, T., Gottlieb. K., Manchester. D. K.. Park, S. S., West, D., Gurtoo,H. L., Tarone. R. E.. and Gelboin, H. V. Monoclonal antibody phenotypingof interindividual differences in cytochrome P-450-dependent reactions ofsingle and twin human placenta. Cancer Res., 44: 3916-3923, 1984.Manchester, D. K.. and Jacoby, E. H. Sensitivity of human placenta!monooxygenase activity to maternal smoking. Clin. Pharmacol. Ther., 30:687-692. 1981.Nowak. D., Schmidt-Preuss. U., Jorres, R., Liebke. F., and Rudiger. H. W.Formation of DNA adducts and water-soluble metabolites ofbenzo|a]pyrenein human monocytes is genetically controlled. Int. J. Cancer. 41: 169-173.1988.Kouri. R. E., McKinney, C. E., Slomiany. D. J.. Snodgrass. D. R.. Wray,N. P., and McLemore, T. L. Positive correlation between high aryl hydrocarbon hydroxylase activity and primary lung cancer as analyzed in cryopre-served lymphocytes. Cancer Res., 42: 5030-5037, 1982.Trell, L.. Korsgaard, R., Janzon, L., and Trell, E. Distribution and repro-ducibility of aryl hydrocarbon hydroxylase inducibility in a prospectivepopulation study of middle-aged male smokers and nonsmokers. Cancer(Phila.), 56: 1988-1994. 1985.Hawke, L. J., and Farrell. G. C. Increased binding of benzo[a]pyrenemetabolites to lymphocytes from patients with lung cancer. Cancer Lett.,JO: 289-297, 1986.Peterson. D. D., McKinney. C. E., Ikeya. K., Smith, H. H.. Bale, A. E.,McBride. O. W.. and Neben, D. W. Human CYPIA1 gene: co-segregationof the enzyme inducible phenotype and a restriction fragment length polymorphism. Am. J. Hum. Genet., 48: 720-70, 1991.Kawajiri. K., Nakachi. K., Imai, K.. Yoshii, A., Shinoda. N.. and Watanabe.J. Identification of genetically high risk individuals to lung cancer by DNApolymorphisms of the cytochrome P450IAI gene. FEBS Lett., 263: 131-133, 1990.Idle. J. R., Mahgoub, A.. Sloan. T. P.. Smith, R. L., Mbanefo. C. O.. andBababunmi, E. A. Some observations on the oxidation phenotype status ofNigerian patients presenting with cancer. Cancer Lett., //: 331-338, 1981.Kaisary, A., Smith, P., Jaczq, E.. McAllister, C. B., Wilkinson, G. R., Ray,W. A., and Branch, R. A. Genetic predisposition to bladder cancer: abilityto hydroxylate debrisoquine and mephenytoin as risk factors. Cancer Res.,47: 5488-5493, 1987.Caporaso, N., Hayes. R. B.. Dosemeci, M.. Hoover, R., Ayesh. R.. Hetzel,M., and Idle. J. R. Lung cancer risk, occupational exposure, and thedebrisoquine metabolic phenotype. Cancer Res., 49: 3675-3679, 1989.Kadlubar, F. F., and Hammons. G. J. The role of cytochrome P450 in themetabolism of chemical carcinogens. In: F. P. Guengerich (ed.). MammalianCytochromes P450. Boca Raton, FL: CRC Press, 1987.Lang, N. P., Chu, D. Z., Hunter, C. F., Kendall, D. C., Flammang, T. J.,and Kadlubar, F. F. Role of aromatic amine acelyltransferase in humancolorectal cancer. Arch. Surg.. 121: 1259-1261, 1986.Gelboin, H. V. Benzo|alpha]pyrene metabolism, activation and carcinogenesis: role and regulation of mixed-function oxidases and related enzymes.Physiol. Rev.. 60: 1107-1166. 1980.Harris. C. C. Concluding remarks: role of carcinogens, cocarcinogens andhost factors in cancer risk. In: C. C. Harris and H. Autrup (eds.). HumanCarcinogenesis. pp. 941-970. New York: Academic Press, 1983.Petruzzelli, S., Camus, A. M., Carrozzi, L., Ghelarducci, L., Rindi, M.,Menconi, G., Angeletti, C. A., Ahotupa. M., Hietanen, E., Aitio, A., Saracci,R., Bartsch, H., and Giuntini, C. Long-lasting effects of tobacco smokingon pulmonary drug-metabolizing enzymes: a case-control study on lungcancer patients. Cancer Res.. 48: 4695-4700, 1988.Jerina. D. M., and Bend. J. A. Glutathione S-transferases. In: D. Jollow, J.J. Kocsis, R. Synder, and H. Vaino (eds.). Biological Reactive Metabolites,pp. 207-236. New York: Plenum Publishing Corp.. 1977.Robertson, I. G. C.. Guthenberg. C.. Mannervik. B.. and Jernstrom. B. Theglutathione conjugation of benzo(a)pyrene diol-epoxide by human glutathi-one transferases. In: M. Cooke and A. J. Dennis (eds.), Polynuclear Aromatic Hydrocarbons: A Decade of Progress, pp. 799-808. Columbus, OH:Battelle Press. 1988.Kelterer. B. Protective role of glutathione and glutathione transferases inmutagenesis and carcinogenesis. Mutât.Res.. 202: 343-361. 1988.Mannervik. B. The isoenzymes of glutathione Iransferase. Adv. Enzymol.,57:357-417, 1985.Seidegard, J.. and Pero, R. W. The hereditary transmission of high glutathione transferase activity towards irans-stilbene oxide in human mononu-clear leukocytes. Hum. Genet., 69: 66-68. 1985.Seidegard, J.. Pero, R. W.. Miller, D. G., and Beattie. E. J. A glutathionetransferase in human leukocytes as a marker for the susceptibility to lungcancer. Carcinogenesis (Lond.). 7: 751-753, 1986.Seidegard, J.. Pero, R. W.. Markowitz, M. M., Roush. G.. Miller, D. G.,and Beattie, E. J. Isoenzyme(s) of glutathione transferase (class Mu) as a

on April 2, 2020. © 1991 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Page 19: Chemical and Physical Carcinogenesis: Advances …...CARCINOGENESIS Table 1 Examples of advances in chemical and physical carcinogenesis" A. Cancer etiology I. Life-style factors (tobacco

CARCINOGENESIS

marker for the susceptibility to lung cancer: a follow up study. Carcinogen-esis (Lond.), //: 33-36. 1990. 285.

260. Daniel. F. B., Stoner. G. D., and Schut, H. A. J. Interindividual variationin the DNA binding of chemical genotoxins following metabolism by humanbladder and bronchus expiants. In: F. J. DeSerres and R. Pero (eds.).Individual Susceptibility to Genotoxic Agents in Human Population, pp. 286.177-199. New York: Plenum Publishing Corp.. 1983.

261. DeFlora. S.. Pelruzzelli. S.. Camoirano, A., Bennicelli. C.. Romano. M..Rindi. M., Ghelarducci. L.. and Giuntini, C. Pulmonary metabolism ofmutagens and its relationship with lung cancer and smoking habits. Cancer 287.Res., 47: 4740-4745. 1987.

262. Harries, G. C., Boobis, A. R.. Collier, N., and Davies, D. S. Interindividual 288.differences in the activation of two hepatic carcinogens to mutagens byhuman liver. Hum. Toxicol.. 5: 21-26, 1986. 289.

263. Minchin. R. F.. McManus. M. E.. Boobis. A. R., Davies, D. S., andThorgeirsson. S. S. Polymorphic metabolism of the carcinogen 2-acetyla-minofluorene in human liver microsomes. Carcinogenesis(Lond.), 6:1721-1724. 1985. 290.

264. Pelkonen. O.. and Neben, D. \V. Metabolism of polycyclic aromatic hydrocarbons: etiologic role in carcinogenesis. Pharmacol. Rev., 34: 189-222,1982. 291.

265. Siegfried. J. M., Rudo, K.. Bryant. B. J.. Ellis. S., Mass, M. J., and Nesnow,S. Metabolism of benzo(a)pyrene in monolayer cultures of human bronchialepithelial cells from a series of donors. Cancer Res., 46: 4368-4371, 1986.

266. Yoo, J. S., Guengerich, F. P., and Yang, C. S. Metabolism of A'-nitrosodi- 292.alk>lamines by human liver microsomes. Cancer Res., 48:1499-1504.1988.

267. Stoner, G. D., and Paolino, M. Human tissue culture systems for studies of 293.carcinogen metabolism and macromolecular interactions. In: J. D. Groop-man and P. L. Skipper (eds.). Molecular Dosimetry and Human Cancer,pp. 105-129. Boca Raton. FL: CRC Press, 1991.

268. Weston. A.. Grover. P. L., and Sims. P. Metabolism and activation ofbenzo[a]pyrene by mouse and rat skin in short-term organ culture and in 294.vivo. Chem-Biol. Interact., 42: 233-250. 1982.

269. Kuroki, T., Hosomi, J., Chida. K.. Hosoi. J.. and Nemolo, N. Interindividualvariations of arylhydrocarbon-hydroxylase activity in cultured human epidermal and dermal cells. Jpn. J. Cancer Res.. 78: 45-53, 1987. 295.

270. Perera. F. P.. Santella. R. M.. Brenner. D., Poirier, M. C., Munshi, A. A.,Fischman. H. K.. and Van Ryzin, J. DNA adducts, protein adducts. andsister chromatid exchange in cigarette smokers and nonsmokers. J. Nati.Cancer Inst. (Bethesda), 79: 449-456, 1987. 296.

271. Vahakangas. K.. Autrup. H.. and Harris. C. C. Interindividual variation incarcinogen metabolism. DNA damage and DNA repair. In: A. Berlin. M.Draper. K. Hemminki. and H. Vainio (eds.). Monitoring Human Exposure 297.to Carcinogenic and Mutagenic Agents, pp. 85-98. Lyon, France: International Agency for Research on Cancer. 1984.

272. Setlow, R. B. Variations in DNA repair among humans. In: C. C. Harrisand H. Autrup (eds.). Human Carcinogenesis, pp. 231-254. New York: 298.Academic Press. Inc.. 1983.

273. Hoeijmakers, J. H. J., and Bootsma, D. Molecular genetics of eukaryoticDNA excision repair. Cancer Cells (Cold Spring Harbor), 2:311 -320. 1990.

274. Lindahl. T., Wood. R. D.. and Karran. P. Molecular deficiencies in human 299.cancer-prone syndromes associated with hypersensitivity to DNA damagingagents. In: J. Brugge, T. Curren, E. Harlow, and F. McCormick (eds.).Origins of Human Cancer. Cold Spring Harbor. NY: Cold Spring Harbor 300.Laboratory Press, in press, 1991.

275. Pero. R. W., Bryngelsson, C., Bryngelsson, T., and Norden, A. A geneticcomponent of the variance of Ar-acetoxy-2-acetylaminofluorene-induced

DNA damage in mononuclear leukocytes determined by a twin study. Hum.Genet., 65: 181-184. 1983. 301.

276. Pero. R. W., Johnson. D. B.. Markowitz. M., Doyle, G., Lund-Pero, M.,Seidegard, J., Halper, M.. and Miller. D. G. DNA repair synthesis inindividuals with and without a family history of cancer. Carcinogenesis 302.(Lond.), 10:693-697, 1989.

277. D'Ambrosio, S. M., Samuel, M. J., Dutta-Choudhury, T. A., and Wani, A.A. O6-methylguanine-DNA methyltransferase in human fetal tissues: fetaland maternal factors. Cancer Res., 47: 51-55. 1987.

278. D'Ambrosio, S. M., Wani. G., Samuel, M., and Gibson-D'Ambrosio. R. E. 303.Repair of O'-methylguanine in human fetal brain and skin cells in culture.Carcinogenesis (Lond.). 5: 1657-1661, 1984.

279. Gerson, S. L., Miller, K.. and Berger. N. A. O6 alkylguanine-DNA alkyl-transferase activity in human myeloid cells. J. Clin. Invest.. 76: 2106-2114,1985.

280. Lawley. P. D., Harris. G.. Phillips. E., Irving. W., Colaco, C. B., Lydyard, 304.P. M., and Roitt, I. M. Repair of chemical carcinogen-induced damage inDNA of human lymphocytes and lymphoid cell lines—studies of the kineticsof removal of O'-methylguanine and 3-methyladenine. Chem.-Biol. Interact.. 57: 107-121, 1986.

281. Grafstrom, R. C., Pegg, A. E., Trump, B. F., and Harris, C. C. O6- 305.Alkylguanine-DNA alkyltransferase activity in normal human tissues andcells. Cancer Res., 44: 2855-2857, 1984.

282. Y'arosh, D. B. The role of O6-methylguanine-DNA methyltransferase in cell 306.

survival, mutagenesis and carcinogenesis. Mutât.Res., 145: 1-16, 1985.283. Myrnes. B.. Giercksky, K. E.. and Krokan, H. Interindividual variation in

the activity of O'-methyl guanine-DNA methyltransferase and uracil-DNAglycosylase in human organs. Carcinogenesis (Lond.), 4: 1565-1568, 1983. 307.

284. Jensen. L.. and Linn. S. A reduced rate of bulky DNA adduci removal iscoincident with differentiation of human neuroblastoma cells induced by

5041s

nerve growth factor. Mol. Cell Biol.. 8: 3964-3968, 1988.Sagher. D.. Karrison. T.. Schwartz, J. L., Larson, R., Meier, P.. and Strauss.B. Low O*-alkylguanine DNA alkyltransferase activity in the peripheralblood lymphocytes of patients with therapy-related acute nonlymphocyticleukemia. Cancer Res.. 48: 3084-3089. 1988.Rudiger, H. W., Schwartz, U.. Serrand, E., Stief. M.. Krause. T., Nowak,D.. Doerjer. G., and Lehnen. G. Reduced O'-methylguanine repair infibroblast cultures from patients with lung cancer. Cancer Res., 49: 5623-5626, 1989.Oesch, F., Aulmann, W., Platt, K. L., and Doerjer, G. Individual differencesin DNA repair capacities in man. Arch. Toxicol. Suppl., 10: 172-179, 1987.Boutwell, R. K. Some biological aspects of skin carcinogenesis. Prog. Exp.Tumor Res., 4: 207-250, 1964.Slaga. T. J., Fischer. S. M., Weeks, C. E., Klein-Szanto, A. J. P., andReiners, J. J. Studies of mechanisms involved in multistage carcinogenesisin mouse skin. In: C. C. Harris and P. A. Cerutti (eds.). Mechanisms ofChemical Carcinogenesis. pp. 207-227. New York: Alan R. Liss, Inc., 1982.Slaga. T. J., and Fischer. S. M. Strain differences and solvent effects inmouse skin carcinogenesis experiments using carcinogens, tumor initiatorsand promoters. Prog. Exp. Tumor Res., 26: 85-109, 1983.Bangrazi, C., Mouton, D., Neveu, T., Sarán,A., Covelli, V., Doria, G., andBiozzi, G. Genetics of chemical carcinogenesis. 1. Bidirectional selectivebreeding of susceptible and resistant lines of mice to two-stage skin carcinogenesis. Carcinogenesis, //: 1711-1719, 1990.Moolgavkar. S. H. Scientific Issues in Quantitative Cancer Risk Assessment.Boston: Birkhauser, 1990.Wogan, G. N. Detection of DNA damage in studies on cancer etiology andprevention. In: H. Bartsch, K. Hemminki, and I. K. O'Neill (eds.). Methodsfor Detecting DNA-Damaging Agents in Humans: Applications in CancerEpidemiology and Prevention. IARC Publication No. 89, pp. 32-51. Lyon,France: International Agency for Research on Cancer, 1988.Sugimura, H.. Weston, A., Caporaso, N. E., Shields. P. G., Bowman, E.D., Metcalf, R. A., and Harris, C. C. Biochemical and molecular epidemiology of cancer. In: L. W. Chang and R. Hart (eds.). Recent Advances inBiomarker Research. New York: Academic Press Inc., in press, 1991.Poirier, M. C. Development of immunoassays for the detection of carcino-gen-DNA adducts. In: P. L. Skipper, F. Koshier. and J. D. Groopman (eds.).Molecular Dosimetry in Human Cancer, pp. 211-229. Boca Raton, FL:CRC Press, 1991.Ehrenberg. L., Hiesche, K. D., Osterman-Golkar, S.. and Wenneberg. I.Evaluation of genetic risks of alkylating agents: tissue doses in the mousefrom air contaminated with ethylene oxide. Mutât.Res., 24: 83-103, 1974.Vineis, P., Caporaso, N.. Tannenbaum, S. R., Skipper, P. L.. Glogowski,J., Bartsch, H.. Coda, M.. Talaska, G.. and Kadlubar. F. Acetylationphenotype, carcinogen-hemoglobin adducts, and cigarette smoking. CancerRes., 50: 3002-3004, 1990.Tornqvist, M., Osterman-Golkar, S., Kautiainen, A., Jensen, S.. Farmer, P.B., and Ehrenberg, L. Tissue doses of ethylene oxide in cigarette smokersdetermined from adduci levels in hemoglobin. Carcinogenesis (Lond.), 7:1519-1521, 1986.Maclure, M., Bryant, M. S.. Skipper, P. L., and Tannenbaum, S. R. Declineof the hemoglobin adduci of 4-aminobiphenyl during withdrawal fromsmoking. Cancer Res.. 50: 181-184, 1990.Farmer, P. B., Bailey. E., Gorf, S. M.. Tornqvist, M., Osterman-Golkar, S.,Kautiainen. A., and Lewis-Enright, D. P. Monitoring human exposure toethylene oxide by the determination of haemoglobin adducts using gaschromatogr;iph>-mass spectrometry. Carcinogenesis (Lond.), 7: 637-640.1986.Wild, C. P.. Jiang, Y. Z.. Allen. S. J.. Hall, A. J., Jansen, L. A. M.. andMontesano, R. Aflatoxin-albumin adducts in human sera from differentregions of the world. Carcinogenesis (Lond.), //: 2271-2274, 1990.Perera, F. P., Poirier, M. C., Yuspa, S. H., Nakayama, J., Jaretzki, A.,Curnen, M. M., Knowles, D. M., and Weinstein, I. B. A pilot project inmolecular cancer epidemiology: determination of benzo[a]pyrene—DNAadducts in animal and human tissues by immunoassays. Carcinogenesis(Lond.). 3: 1405-1410, 1982.Haugen, A., Bêcher,G., Benestad, C., Vahakangas, K., Trivers, G. E.,Newman, M. J., and Harris, C. C. Determination of polycyclic aromatichydrocarbons in the urine, benzo(a)pyrene diol epoxide-DNA adducts inlymphocyte DNA. and antibodies to the adducts in sera from coke ovenworkers exposed to measured amounts of polycyclic aromatic hydrocarbonsin the work atmosphere. Cancer Res., 46: 4178-4183. 1986.Harris. C. C.. Vahakangas. K.. Newman, M. J., Trivers, G. E., Shamsuddin.A. K. M., Sinopoli, N. T., Mann, D. L., and Wright, W. E. Detection ofbenzo[o]pyrene diol epoxide-DNA adducts in peripheral blood lymphocytesand antibodies to the adducts in serum from coke oven workers. Proc. Nail.Acad. Sci. USA, 82: 6672-6676, 1985.Shamsuddin, A. K. M., Sinopoli, N. T., Hemminki. K., Boesch, R. R., andHarris, C. C. Detection of benzo(a)pyrene:DNA adducts in human whiteblood cells. Cancer Res., 45: 66-68, 1985.Liou, S. H., Jacobson-Kram. D., Poirier, M. C., Nguyen, D.. Strickland, P.T.. and Tockman, M. S. Biological monitoring of fire fighters: sister chromatid exchange and polycyclic aromatic hydrocarbon-DNA adducts in peripheral blood cells. Cancer Res.. 49: 4929-4935, 1989.Strickland. P. T., Rothman, N.. Baser. M. E., and Poirier, M. C. Polycyclicaromatic hydrocarbon-DNA adduci load in peripheral blood cells: contribution of multiple exposure sources. In: M. Vanderlaan, L. H. Stanker, B.

on April 2, 2020. © 1991 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Page 20: Chemical and Physical Carcinogenesis: Advances …...CARCINOGENESIS Table 1 Examples of advances in chemical and physical carcinogenesis" A. Cancer etiology I. Life-style factors (tobacco

CARCINOGENESIS

E. Walkins, and D. W. Roberts (eds.). Immunoassays for Trace Chemical 330.Analysis. Monitoring Toxic Chemicals in Humans. Food, and the Environment, pp. 257-263. Washington, DC: American Chemical Society, 1991. 331.

308. Hemminki, K., Grzybowska, E., Chorazy, M., Twardowska-Sauch, K.,Sroczynski, J. W., Putnam, K. L., Randerath. K., Phillips, D. H., Hewer, 332.A., Santella, R. M., and Perera, F. P. DNA adducts in humans related tooccupational and environmental exposure to aromatic compounds. IARC 333.Publication. Lyon. France: International Agency for Research on Cancer,in press, 1991. 334.

309. Newman. M. J., Light, B. A.. Weston. A.. Tollurud, D., Clark, J. L., Mann,D. L., Blackmon, J. P., and Harris, C. C. Detection and characterization of 335.human serum antibodies to polycyclic aromatic hydrocarbon diol-epoxideDNA adducts. J. Clin. Invest., «2:145-153, 1988. 336.

310. Weston. A., Rowe, M. L., Manchester. D. K., Farmer, P. B., Mann, D. L.,and Harris, C. C. Fluorescence and mass spectral evidence for the formation 337.of benzo[a]pyrene onf/'-diol-epoxide-DNA and -hemoglobin adducts in hu

mans. Carcinogenesis (Lond.j, 10: 251-257. 1989. 338.311. Manchester, D. K., Wilson, V. L., Hsu, I. C., Choi, J. S., Parker, N. B.,

Mann, D. L., Weston, A., and Harris, C. C. Synchronous fluorescencespectroscopic, immunoaffmity Chromatographie, and 32P-postlabeling analy- 339.

sis of human placenta! DNA known to contain benzo(a)pyrene dial epoxideadducts. Carcinogenesis (Lond.), //: 553-559. 1990. 340.

312. Manchester, D. K., Weston, A., Choi, J. S., Trivers, G. E., Fennessey, P.,Quintana, E., Farmer, P. B., Mann, D. L., and Harris. C. C. Detection ofbenzo[a]pyrene diol epoxide-DNA adducts in human placenta. Proc. Nati. 341.Acad. Sci. USA, 85: 9243-9247, 1988.

313. Bowman, E. D., Shields, P. G., Poirier, M. C., Santella, R. M., Manchester, 342.D. K., Farmer, P. B., Harris, C. C., and Weston, A. Determination ofcomplete fluorescence excitation-emission matrices for polycyclic aromatichydrocarbon-DNA adducts found in human lung and placenta. Proc. Am. 343.Assoc. Cancer Res., 31: 89, 1990.

314. Weston. A., Manchester, D. K., Poirier, M. C., Choi, J. S., Trivers, G. E.,Mann, D. L., and Harris, C. C. Derivative fluorescence spectral analysis of 344.polycyclic aromatic hydrocarbon-DNA adducts in human placenta. Chem.Res. Toxicol., 2: 104-108, 1989. 345.

315. Everson, R. B., Randerath, E., Santella, R. M., Avitts, T. A., Weinstein, I.B.. and Randerath. K. Quantitative associations between DNA damage in 346.human placenta and maternal smoking and birth weight. J. Nati. CancerInst., 80: 567-576. 1988.

316. Phillips, D. H., Hemminki, K., Alhonen, A., Hewer, A., and Grover, P. L. 347.Monitoring occupational exposure to carcinogens: detection by 32P-postla-belling of aromatic DNA adducts in white blood cells from iron foundry 348.workers. Mutât.Res., 204: 531-541, 1988.

317. Herrón, D. C., and Shank, R. C. Methylated purines in human liver DNA 349.after probable dimethylnitrosamine poisoning. Cancer Res., 40:3116-3117,1980.

318. Umbenhauer, D.. Wild, C. P., Montesano, R., Saffhill. R., Boyle, J. M., 350.Huh, N.. Kirstein, U., Thomale, J., Rajewsky, M. F., and Lu, S. H. 0'-

methyldeoxyguanosine in oesophageal DNA among individuals at high risk 351.of oesophageal cancer. Int. J. Cancer, 36: 661-665, 1985.

319. Saffhill, R., Badawi, A. F., and Hall, C. N. Detection of 06-methylguaninein human DNA. In: H. Bartsch, K. Hemminki, and I. K. O'Neill (eds.), 352.

Methods for Detecting DNA Damaging Agents in Humans: Applicationsin Cancer Epidemiology and Prevention, pp. 301-305. Lyon, France: Inter- 353.national Agency for Research on Cancer, 1988.

320. Wilson, V. L., Weston, A., Manchester, D. K., Trivers, G. E., Roberts, D.W., Kadlubar, F. F., Wild, C. P., Montesano, R., Willey, J. C., Mann, D. 354.L., and Harris, C. C. Alkyl and aryl carcinogen adducts detected in humanperipheral lung. Carcinogenesis (Lond.), 10: 2149-2153, 1989. 355.

321. Foiles, P. G., Miglietta, L. M., Akerkar, S. A., Everson, R. B., and Hecht,S. S. Detection of O'-methyldeoxyguanosine in human placental DNA.Cancer Res., 48: 4184-4188, 1988. 356.

322. Huh, N. H., Satoh, M. S., Shiga, J., Rajewsky, M. F., and Kuroki, T.Immunoanalytical detection of O4-ethylthymine in liver DNA of individualswith or without malignant tumors. Cancer Res., 49: 93-97, 1989.

323. Wilson, V. L., Basu, A. K., Essigmann, J. M., Smith, R. A., and Harris, C. 357.C. O'-alkyldeoxyguanosine detection by 32P-postlabeling and nucleotideChromatographie analysis. Cancer Res., 48: 2156-2161, 1988.

324. Wilson, V. L., Masui, T., Smith, R. A., and Harris, C. C Genomic 5- 358.methyldeoxycytidine decreases associated with the induction of squamousdifferentiation in cultured normal human bronchial epithelial cells. Carci- 359.nogenesis (Lond.), 9: 2155-2159, 1988.

325. Shields, P. G., Povey, A. C., Wilson, V. L., and Harris, C. C. 32P-postla- 360.belling of jVmethyldeoxyguanosine (A"medG). Proc. Am. Assoc. Cancer

Res., 50:316, 1989. 361.326. Shields, P. G., Povey, A. C., Wilson, V. L., Weston, A., and Harris, C. C.

Combined high performance liquid chromatography/"P-postlabeling assay 362.of A^-methyldeoxyguanosine. Cancer Res., 50: 6580-6584. 1990.

327. Bennett. W. P., Hollstein, M. C., He, A., Zhu, S. M., Resau, J., Trump, B.F., Metcalf, R. A., Welsh, J. A., Gannon, J. V.. Lane. D. P., and Harris, C. 363.C. Archival analysis of p53 genetic and protein alterations in Chineseesophageal cancer. Oncogene, in press, 1991. 364.

328. Proceedings of the American Academy of Arts and Sciences. Daedalus, Fall1990, "Risk". Cambridge. MA: American Academy of Arts and Sciences,

1990. 365.329. Hill, J. Cautions against the immoderate use of snuff. London: R. Baldwin

and J. Jacobson, 1761.

5042s

Soemmerring, S. T. De Morbis Pasorum Absorbeutium Corporis Humani.Frankfurtam Main, Germany: Varrentrapp & Weuner, 1795.Doll, R., and Hill, A. B. Smoking and carcinoma of the lung: preliminaryreport. Br. Med. J., 2: 739-748, 1950.Wynder, E. L., and Graham, E. A. Tobacco smoking as a possible etiologicfactor in bronchogenic carcinoma. JAMA, 143: 329-336, 1950.Rama/vini. B. De Morbis Artificum, Diatriba, (Diseases of Workers). NewYork: Hafner. 1964.Pott, P. Chirurgical observations relative to the cancer of the scrotum.London: Hawes, L., Clark, W.. and Collins, R., 1775.Thiersch, K. Der Epithelialkrebs, Namentlieh der Ausseren Haut. Germany:Leipzig, 1875.Harting, F. H.. and Hesse, W. Der lungenkrebs, die bergkrankheit in denschneeberger gruben. Vrtljschr. Gerlichtl. Med., 30: 296-309, 1879.Renn, L. Blasengeschwulste bei fuchsin-arbeiten. Arch. Klin. Chir., SO:588-600, 1895.Frieben, A. Demonstration lines cancroids des rechten Handrückens,dassich nach langdauernder Einwirkung von Rontgenstrahlen entwichelt hatte.Fortschr. Geb. Rontgenstr., 6: 106-117, 1902.Clunet, J. Recherches expérimentalessur les tumeurs malignes. Paris:Steinheil. 1910.Delore. P.. and Borgomano, C. Leucemie alignéeau cours de l'intoxicationbenzenique: sur l'origine toxique de certaines leucémiesaiguëset leur

relations avec les anémiesgraves. J. Med. Lyon, 9: 227, 1928.Martland, H. S., and Humphries, R. E. Osteogenic sarcoma in dial paintersusing luminous paint. Arch. Pathol.. 7: 406-417, 1929.Minister of Labour and National Service. Annual Report of the ChiefInspector of Factories for the Year 1947 (CMD. 7621). London: HisMajesty's Stationery Office, 1949.Wagner, J. C., Sleggs, C. A., and Marchand, P. Diffuse pleural meso-thelioma and asbestos exposure in the North Western Cape Province. Br.J. Ind. Med., 17: 260-271, 1960.Yamagiwa, K., and Ichikawa, K. Experimental study of the pathogenesis ofcarcinoma. J. Cancer Res., J: 1-21, 1918.Kennaway, E. L., and Hieger, I. Carcinogenic substances and their fluorescence spectra. Br. Med. J.. /: 1044-1046, 1930.Hueper, W. C., Wiley, F. H., and Wolfe, H. D. Experimental productionof bladder tumors in dogs by administration of betanaphthylamine. J.Industr. Hyg., 20:46-84, 1938.Turner, F. C. Sarcomas at sites of subcutaneously implanted Bakelite disksin rats. J. Nat!. Cancer. Inst. (Bethesda), 2: 81-83, 1941.Berenblum, I. The cocarcinogenic action of croton resin. Cancer Res., /:44-48, 1941.Fujiki, H., Mori, M., Nakayasu, M., Terada, M., and Sugimura, T. Apossible naturally occurring tumor promoter, teleocidin B from Streplo-myces. Biochem. Biophys. Res. Commun., 90: 976-983, 1979.Triólo,V. A. Nineteenth century' foundations of cancer research: origins ofexperimental research. Cancer Res., 24: 4-27, 1964.Triólo, V. A. Nineteenth century foundations of cancer research: advancesin tumor pathology, nomenclature and theories of oncogenesis. Cancer Res.,25:75-106, 1965.Shimkin, M. B. Some Classics of Experimental Oncology. Washington,DC: Government Printing Office, 1980.Upton, A. C. Physical Carcinogenesis: radiation—history and sources. In:D. Becker (ed.). Cancer: A Comprehensive Treatise, Ed. 2, pp. 551-567.New York: Plenum Publishing Corp., 1982.Witkowski, J. A. The inherited character of cancer—an historical survey.Cancer Cells (Cold Spring Harbor). 2: 228-257, 1990.Cook, J. W., Duffy, E., and Schoental, R. Primary liver tumours in ratsfollowing feeding with alkaloids of Senecio jacobaea. Br. J. Cancer, 4: 405-410, 1950.Schoental, R., Head, M. A., and Peacock, P. R. Senecio alkaloids: primaryliver tumors in rats as a result of treatment with (1) a mixture of alkaloidsfrom S. jacobaea Linn., (2) retrorsine, and (3) isatidine. Br. J. Cancer, 8:458-465, 1954.Laqueur, G. L. Carcinogenic effects of cycad meal and cycasin, methylaz-oxymethanol glycoside, in rats and effects of cycasin in germfree rats. Fed.Proc., 23: 1386-1387. 1964.Evans, I. A., and Mason, J. Carcinogenic activity of bracken. Nature (Lond.),20«:913-914, 1965.Carnaghan, R. B. A. Hepatic tumours in ducks fed a low level of toxicgroundnut meal. Nature (Lond.), 20«:308, 1965.Wogan, G. N. Chemical nature and biological effects of the aflatoxins.Bacteriol. Rev., 30: 460-470, 1966.Wogan, G. N., and Newberne, P. M. Dose-response characteristics ofaflatoxin Bl Carcinogenesis in the rat. Cancer Res., 27: 2370-2376, 1967.Magee, P. N., and Barnes, J. M. The production of malignant primaryhepatic tumours in the rat by feeding dimethylnitrosamine. Br. J. Cancer,10: 114-122, 1956.Kuratsune, M. Benzo[a]pyrene content of certain pyrogenic materials. J.Nati. Cancer Inst. (Bethesda), 16: 1485-1496. 1956.Lijinsky, W., and Shubik. P. Benzo[o|pyrene and other polynuclear hydrocarbons in charcoal-broiled meat. Science (Washington DC), 145: 53-55,1964.Sugimura, T., Kawachi, T., Nagao, M., Yahagi, T., Seino, Y., Okamoto,T., Shudo, K., Kosuge, T., Tsuji, K., Wakabayashi, K., litaka, Y., and Hai,A. Mutagenic principle(s) in tryptophan and phenylalanine pyrolysis prod-

on April 2, 2020. © 1991 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Page 21: Chemical and Physical Carcinogenesis: Advances …...CARCINOGENESIS Table 1 Examples of advances in chemical and physical carcinogenesis" A. Cancer etiology I. Life-style factors (tobacco

CARCINOGENESIS

ucts. Proc. Jpn. Acad., 53: 58-61.1977. 396.366. Rasai, H.. Yamaizumi. Z., Wakabayashi. K., Nagao. M.. Sugimura. T.,

Yokoyama. S., Miyazawa, T., Spingarn, N. E., Weisburger, J. H., and 397.Nishimura, S. Potent novel mutagens produced by broiling fish undernormal conditions. Proc. Jpn. Acad., 56B: 278-283, 1980. 398.

367. Wynder, E. L.. Bross, I. J., and Feldman, R. M. A study of the etiologicalfactors in cancer of the mouth. Cancer (Phila.). 10: 1300-1323, 1957. 399.

368. Keller, A. Z. Cirrhosis of the liver, alcoholism and heavy smoking associatedwith cancer of the mouth and pharynx. Cancer (Phila.), 20: 1015-1022, 400.1967.

369. Rothman. K., and Keller, A. The effect of joint exposure to alcohol andtobacco on risk of cancer of the mouth and pharynx. J. Chronic Dis., 25: 401.711-716. 1972.

370. Stewart, A.. Webb, J.. and Hewitt, D. A survey of childhood malignancies. 402.Br. Med. J., /: 1495-1508. 1958.

371. Selikoff, I. J., Hammond, E. C., and Churg, J. Asbestos exposure, smoking, 403.and neoplasia. JAMA, 204: 106-112, 1968.

372. Alpert. M. E., Huit. M. S.. and Davidson, C. S. Hepatoma in Uganda. A 404.study in geographic pathology. Lancet, /: 1265-1267. 1968.

373. Van Duuren. B. L., Goldschmidt. B. M., Langseth, L., Mercado, G., and 405.Sivak. A. n-Haloethers: a new type of alkylating carcinogen. Arch. Environ.Health, 16:472-476, 1968. 406.

374. Viola, P. L., Bigotti, A., and Caputo, A. Oncogenic response of rat skin,lungs, and bones to vinyl chloride. Cancer Res., }/: 516-522, 1971.375. Herbst. A. L., Cole, P.".Colton, T., Robboy, S. J., and Scully, R. E. Age- 407.

incidence and risk of diethylstilbestrol-related clear cell adenocarcinoma ofthe vagina and cervix. Am. J. Obstet. Gynecol., 128: 43-50. 1977. 408.

376. Stanton, M. F., Layard, M., Tegeris, A., Miller, E., May, M.. Morgan, E.,and Smith, A. Relation of particle dimension to carcinogenicity in amphiboleasbestoses and other fibrous minerals. J. Nati. Cancer Inst. (Bethesda), 67: 409.965-975. 1981.

377. Sasaki, T., and Yoshida, T. Liver carcinoma induced by feeding O-amido- 410.azotoluene. Virchows Arch. Pathol. Anat., 295: 175-220, 1935.

378. Boyland, E. Metabolism of polycyclic compounds. Production of dihydrox- 411.ydihydroanthracene from anthracene. Biochem. J., 29: 2679-2683, 1935.

379. Rous. P., and Kidd, J. G. Conditional neoplasms and subthreshold neoplas-tic states. J. Exp. Med., 73: 365-389. 1941.

380. Mottram, J. C. A developing factor in experimental blastogenesis. J. Pathol. 412.Bacterial., 56: 181-187, 1944.

381. Berenblum. I., and Shubik, P. An experimental study of the initiating state 413.of carcinogenesis. and a re-examination of the somatic cell theory of cancer.Br. J. Cancer, 3: 109-118. 1949.

382. Miller, E. C., and Miller, J. A. The presence and significance of bound 414.aminoazo dyes in the livers of rats fed p-dimethylaminoazobenzene. CancerRes., 7:468-480, 1947.

383. Miller, E. C., Miller, J. A., and Enomoto, M. The comparative carcinogen-icities of 2-acetylaminofluorene and its ¿V-hydroxymetabolite in mice. 415.hamsters, and guinea pigs. Cancer Res., 24: 2018-2032, 1964.

384. Murray, A. W., and Fitzgerald, D. J. Tumor promoters inhibit metaboliccooperation in cocultures of epidermal and 3T3 cells. Biochem. Biophys.Res. Commun., 91: 395-401, 1979. 416.

385. Yotti, L. P.. Chang, C. C., and Trosko, J. E. Elimination of metaboliccooperation in Chinese hamster cells by a tumor promoter. Science (Wash- 417ington DC). 206: 1089-1091. 1979.

386. Driedger, P. E.. and Blumberg, P. M. Specific binding of phorbol estertumor promoters. Proc. Nati. Acad. Sci. USA, 77: 567-571, 1980.

387. Castagna, M., Takai, Y., Kaibuchi, K., Sano, K., Kikkawa, U., and Nishi- 418zuka, Y. Direct activation of calcium-activated, phospholipid-dependentprotein kinase by tumor-promoting phorbol esters. J. Biol. Chem., 257:7847-7851, 1982. 419

388. Land, H.. Parada, L. F., and Weinberg, R. A. Tumorigenic conversion ofprimary embryo fibroblasts requires at least two cooperating oncogenes.Nature (Lond.), 304: 596-602, 1983. 420

389. Ruley, H. E. Adenovirus early region 1A enables viral and cellular transforming genes to transform primary cells in culture. Nature (Lond.). 304: 421.602-606, 1983.

390. Davis, R. J., and Czech, M. P. Tumor-promoting phorbol diesters cause thephosphorylation of epidermal growth factor receptors in normal human 422fibroblasts at threonine-654. Proc. Nati. Acad. Sci. USA. 82: 1974-1978,1985.

391. Arcoleo, J. P., and Weinstein. I. B. Activation of protein kinase C by tumor 423promoting phorbol esters, teleocidin and aplysiatoxin in the absence ofadded calcium. Carcinogenesis (Lond.), 6: 213-217, 1985.

392. Ebeling, J. G., Vandenbark, G. R., Kühn,L. J., Ganong, B. R., Bell, R. M.,and Niedel, J. E. Diacylglycerols mimic phorbol diester induction of leu- 424kemic cell differentiation. Proc. Nati. Acad. Sci. USA, 82: 815-819. 1985.

393. Housey, G. M., Johnson. M. D., Hsiao, W. L.. O'Brian. C. A., Murphy, J.

P.. Kirschmeier, P., and Weinstein, I. B. Overproduction of protein kinaseC causes disordered growth control in rat fibroblasts. Cell, 52: 343-354, 4251988.

394. Persons, D. A., Wilkison, W. O., Bell, R. M., and Finn, O. J. Alteredgrowth regulation and enhanced tumorigenicity of NIH 3T3 fibroblasts 426transfected with protein kinase C-I cDNA. Cell, 52: 447-458. 1988.

395. Vogelstein, B., Fearon, E. R., Hamilton, S. R., Kern. S. E., Preisinger, A.C.. Leppert, M., Nakamura, Y., White, R., Smits, A. M.. and Bos, J. L.Genetic alterations during colorectal-tumor development. N. Engl. J. Med., 427319: 525-532, 1988.

5043s

Watkins, D. J. G. A family tree bearing upon the question of the inheritanceof cancer. Br. Med. J.. /: 190, 1904.Boveri, T. Zur Frage der Entstehung Maligner Tumoren, Vol. 1. Jena:Gustave Fischer Verlag, 1914.Muller, H. J. Artificial transmutation of the gene. Science (WashingtonDC), 66; 84-87, 1928.Furth, J., Seibold. H. R.. and Rathbone, R. R. Experimental studies onlymphomatosis of mice. Am. J. Cancer, 19: 521-604, 1933.Strong, L. C. The establishment of the C3H inbred strain of mice for thestudy of spontaneous carcinoma of the mammary gland. Genetics. 20: 586-591, 1935.Charles, D. R., and Luce-Clausen, E. M. The kinetics of papilloma formation in benzpyrene-treated mice. Cancer Res., 2: 261, 1942.Comings, D. E. A general theory of carcinogenesis. Proc. Nati. Acad. Sci.USA, 70:3324-3328, 1973.Auerbach, C., Robson, J. M., and Carr, J. G. The chemical production ofmutations. Science (Washington DC), 105: 243-247, 1947.Boyland, E., and Horning, E. S. The induction of tumours with nitrogenmustards. Br. J. Cancer, 3: 118-123, 1949.Boyland, E. Different types of carcinogens and their possible modes ofaction. Cancer Res., 12: 77-84, 1952.Wheeler, G. P., and Skipper, H. E. Studies with mustards. III. In vivofixation of C'4Hj in nucleic acid fractions of animal tissues. Arch. Biochem.Biophys., 72:465-475, 1957.Brookes, P., and Lawley, P. D. The reaction of mustard gas with nucleicacids in vitro and in vivo. Biochem. J., 77:478-484, 1960.Farber, E., and Magee, P. N. The probable alkylation of liver ribonucleicacid by the hepatic carcinogens dimethylnitrosamine and ethionine.Biochem. J.. 76: 58P. 1960.Nowell, P. C., and Hungerford, D. A. A minute chromosome in humangranulocytic leukemia. Science (Washington DC), 132: 1497, 1960.Beukers, R.. and Berends. W. Isolation and identification of the irradiationproduct of thymine. Biochim. Biophys. Acta, 41: 550-551, 1960.Brookes, P.. and Lawley, P. D. Evidence for the binding of polynucleararomatic hydrocarbons to the nucleic acids of mouse skin: relation betweencarcinogenic power of hydrocarbons and their binding to deoxyribonucleicacid. Nature (Lond.), 202: 781-784, 1964.Cleaver, J. E. Defective repair replication of DNA in xeroderma pigmen-tosum. Nature (Lond.), 218: 652-656, 1968.Kuntzman, R., Mark, L. C., Brand, L.. Jacobson, M., Levin, W., andConney, A. H. Metabolism of drugs and carcinogens by human liver enzymes. J. Pharmacol. Exp. Ther.. 152: 151-156. 1966.Welch, R. M., Harrison, Y. E., Conney, A. H., Poppers, P. J., and Finster,M. Cigarette smoking: stimulatory effect on metabolism of 3,4-benzpyreneby enzymes in human placenta. Science (Washington DC), 760: 541-542,1968.Harris, C. C., Autrup. H., Connor, R. D., Barrett, L. A., McDowell, E. M.,and Trump, B. F. Interindividual variation in binding of benzo[a]pyrene toDNA in cultured human bronchi. Science (Washington DC), 194: 1067-1069, 1976.Harris, H., Miller, O. J., Klein, G., Worst, P., and Tachibana, T. Suppression of malignancy by cell fusion. Nature (Lond.), 223: 363-368, 1969.Ames, B. N., Durston, W. E., Yamasaki. E.. and Lee. F. D. Carcinogensare mutagens: a simple test system combining liver homogenates for activation and bacteria for detection. Proc. Nati. Acad. Sci. USA, 70: 2281-2285, 1973.McCann, J., Choi, E., Yamasaki. E., and Ames, B. N. Detection of carcinogens as mutagens in the Salmonella/microtome test: assay of 300 chemicals. Proc. Nati. Acad. Sci. USA, 72: 5135-5139, 1975.Stehelin, D., Varmus, H. E., Bishop, J. M., and Vogt, P. K. DNA relatedto the transforming gene(s) of avian sarcoma viruses is present in normalavian DNA. Nature (Lond.), 260: 170-173, 1976.de Larco, J. E., and Todaro, G. J. Growth factors from murine sarcomavirus-transformed cells. Proc. Nati. Acad. Sci. USA, 75:4001-4005, 1978.Shih, C., Shilo, B. Z., Goldfarb, M. P., Dannenberg, A., and Weinberg, R.A. Passage of phenotypes of chemically transformed cells via transfectionof DNA and chromatin. Proc. Nati. Acad. Sci. USA, 76: 5714-5718, 1979.Stanbridge, E. J., Flandermeyer, R. R., Daniels, D. W., and Nelson-Rees,W. A. Specific chromosome loss associated with the expression of tumorigenicity in human cell hybrids. Somatic Cell. Genet., 7: 699-712, 1981.Dalla-Favera, R.. Bregni, M.. Erikson, J., Patterson, D., Gallo, R. C., andCroce, C. M. Human c-myc one gene is located on the region of chromosome8 that is translocated in Burkitt lymphoma cells. Proc. Nati. Acad. Sci.USA, 79:7824-7827, 1982.De Klein, A., Van Kessel, A. G., Grosveld, G., Bartram, C. R., Hagemeijer,A.. Bootsma, D., Spurr, N. K.. Heisterkamp, N., Groffen, J., and Stephen-son. J. R. A cellular oncogene is translocated to the Philadelphia chromosome in chronic myelocytic leukaemia. Nature (Lond.). 300:765-767, 1982.Shen-Ong, G. L.. Keath, E. J., Piccoli, S. P., and Cole, M. D. Novel myconcogene RNA from abortive immunoglobulin-gene recombination inmouse plasmacytomas. Cell, 31: 443-452, 1982.Taub, R.. Kirsch, L, Morton. C., Lenoir, G., Swan, D., Tronick, S., Aar-onson, S., and Leder, P. Translocation of the c-myc gene into the immu-noglobulin heavy chain locus in human Burkitt lymphoma and murineplasmacytoma cells. Proc. Nati. Acad. Sci. USA, 79: 7837-7841, 1982.Goldfarb, M., Shimizu, K.. Perucho, M., and Wigler, M. Isolation andpreliminary characterization of a human transforming gene from T24 blad-

on April 2, 2020. © 1991 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Page 22: Chemical and Physical Carcinogenesis: Advances …...CARCINOGENESIS Table 1 Examples of advances in chemical and physical carcinogenesis" A. Cancer etiology I. Life-style factors (tobacco

CARCINOGENESIS

der carcinoma cells. Nature (Lond.), 296:404-409, 1982.428. Pulciani. S., Santos, E.. Lauver, A. V.. Long, L. K., Robbins. K. C., and

Barbacid, M. Oncogenes in human tumor cell lines: molecular cloning of atransforming gene from human bladder carcinoma cells. Proc. Nati. Acad.Sci. USA. 79:2845-2849. 1982.

429. Shih, C.. and Weinberg. R. A. Isolation of a transforming sequence from ahuman bladder carcinoma cell line. Cell. 29: 161-169. 1982.

430. Reddy, E. P., Reynolds, Santos, E.. and Barbacid, M. A point mutation isresponsible for the acquisition of transforming properties by the T24 humanbladder carcinoma oncogene. Nature (Lond.), 300: 149-152, 1982.

431. Tabin, C. J., Bradley, S. M., Bargmann, C. 1., Weinberg, R. A., Papageorge,A. G., Scolnick. E. M., Dhar, R., Lowy, D. R., and Chang, E. H. Mechanismof activation of a human oncogene. Nature (Lond.), 300: 143-149, 1982.

432. Taparowsky, E.. Suard, V'., Fasano, O., Shimizu, K., Goldfarb, M., and

Wigler, M. Activation of the T24 bladder carcinoma transforming gene islinked to a single amino acid change. Nature (Lond.), 300: 762-765, 1982.

433. Balmain. A., and Pragnell. I. B. Mouse skin carcinomas induced in vivo bychemical carcinogens have a transforming Harvey-raj oncogene. Nature(Lond.). 303: 72-74, 1983.

434. Sukumar, S., Notario, V.. Martin-Zanca. D., and Barbacid, M. Inductionof mammary carcinomas in rats by nitroso-methylurea involves malignantactivation of H-ras-1 locus by single point mutations. Nature (Lond.), 306:658-661. 1983.

435. Downward. J.. Yarden, Y., Mayes, E., Scrace, G., Totty, N., Stockwell, P.,Ullrich, A., Schlessinger. J., and Waterfield, M. D. Close similarity ofepidermal growth factor receptor and \-erb-B oncogene protein sequences.Nature (Lond.). 307: 521-527. 1984.

436. Preston. B. D., Singer, B.. and Loeb. L. A. Mutagenic potential of O4-methylthymine in vivo determined by an enzymatic approach to site-specificmutagenesis. Proc. Nati. Acad. Sci. USA, 83: 8501-8505. 1986.

437. Yoakum, G. H., Lechner, J. F., Gabrielson. E. W., Korba, B. E., Malan-Shibley. L., Willey, J. C, VaMio, M. G., Shamsuddin, A. K. M., Trump,B. F., and Harris, C. C. Transformation of human bronchial epithelial cellstransfected by Harvey ras oncogene. Science (Washington DC), 227: 1174-1179. 1985.

438. Rhim. J. S., Jay, G., Arnstein, P., Price, F. M., Sanford. K. K., andAaronson, S. A. Neoplastic transformation of human epidermal keratino-cytes by AD12-SV40 and Kirsten sarcoma viruses. Science (WashingtonDC), 227:1250-1252, 1985.

439. Friend, S. H., Bernards, R., Rogelj, S., Weinberg, R. A., Rapaport, J. M.,Albert, D. M., and Dryja, T. P. A human DNA segment with properties ofthe gene that predisposes to retinoblastoma and osteosarcoma. Nature(Lond.), 323: 643-646, 1986.

440. Maki, Y., Bos, T. J., Davis, C., Starbuck, M., and Vogt, P. K. Aviansarcoma virus 17 carries thej'wn oncogene. Proc. Nati. Acad. Sci. USA, 84:

2848-2852, 1987.441. Vogt. P. K., Bos. T. J., and Doolittle, R. F. Homology between the DNA-

binding domain of the GCN4 regulatory protein of yeast and the carboxyl-terminal region of a protein coded for by the oncogene jun. Proc. Nati.Acad. Sci. USA, 84: 3316-3319, 1987.

442. Bohmann, D., Bos, T. J., Admon, A., Nishimura, T., Vogt, P. K., and Tjian,R. Human proto-oncogene c-jun encodes a DNA binding protein withstructural and functional properties of transcription factor AP-1. Science(Washington DC). 238: 1386-1392, 1987.

443. Baker, S. J.. Fearon, E. R., Nigro, J. M., Hamilton. S. R., Preisinger, A.C., Jessup. J. M., vanTuinen, P.. Ledbetter. D. H., Barker, D. F., andNakamura, Y. Chromosome 17 deletions and p53 gene mutations in colo-rectal carcinomas. Science (Washington DC). 244: 217-221, 1989.

444. Eliyahu, D., Michalovitz, D., Eliyahu. S., Pinhasi-Kimhi, O., and Oren, M.Wild-type p53 can inhibit oncogene-mediated focus formation. Proc. Nati.Acad. Sci. USA, 86: 8763-8767. 1989.

445. Finlay, C. A., Hinds, P. W., and Levine, A. J. The p53 proto-oncogene canact as a suppressor of transformation. Cell, 57: 1083-1093, 1989.

446. Fearon, E. R.. Cho, K. R.. Nigro, J. M., Kern, S. E., Simons, J. W.,Ruppert, J. M., Hamilton, S. R., Preisinger, A. C., Thomas, G., Kinzler, K.W.. and Vogelstein. B. Identification of a chromosome 18q gene that isaltered in colorectal cancers. Science (Washington DC), 2^7:49-56. 1990.

447. Cawthon, R. M.. Weiss, R., Xu, G. F., Viskochil, D.. Culver, M., Stevens,J., Robertson, M., Dunn, D., Gesteland. R., O'Connell, P., and White. R.

A major segment of the neurofibromatosis type 1 gene: cDNA sequence,genomic structure, and point mutations (published erratum appears in Cell62(3): following 608, 1990]. Cell, 62: 193-201, 1990.

448. Xu. G. F., O'Connell, P., Viskochil, D., Cawthon. R.. Robertson, M.,

Culver, M.. Dunn, D., Stevens, J., Gesteland, R., White, R., and Weiss, R.The neurofibromatosis type 1 gene encodes a protein related to GAP. Cell,«2:599-608, 1990.

449. Wallace, M. R., Marchuk, D. A.. Andersen, L. B.. Letcher. R., Odeh. H.M., Saulino, A. M., Fountain, J. W., Brereton, A.. Nicholson. J., Mitchell,A. L., Brownstein, B. H.. and Collins, F. S. Type 1 neurofibromatosis gene:identification of a large transcript disrupted in three NF1 patients. Science(Washington DC), 249: 181-186, 1990.

450. Call, K. M., Glaser, T.. Ito, C. Y., Buckler, A. J.. Pelletier, J., Haber, D.

A.. Rose, E. A., Krai, A., Yeger, H., Lewis, W. H., Jones, C., and Housman,D. E. Isolation and characterization of a zinc finger polypeptide gene at thehuman chromosome 11 Wilms' tumor locus. Cell, 60: 509-520, 1990.

451. Gessler, M., Poustka, A., Cavenee. W., Neve, R. L., Orkin, S. H., andBruns, G. A. Homozygous deletion in Wilms tumours of a zinc-finger geneidentified by chromosome jumping. Nature (Lond.). 343: 774-778, 1990.

452. Kinzler, K. W., Nilbert. M. C., Vogelstein, B., Bryan, T. M., Levy, D. B.,Smith, K. J., Preisinger, A. C., Hamilton, S. R., Hedge. P., Markham, A.,Carlson, M., Joslyn, G., Groden, J., White, R., Miki, Y., Miyoshi, Y.,Nishisho, L, and Nakamura, Y. Identification of a chromosome 5q21 genethat is mutated in colorectal cancers. Science (Washington DC), 251: 1366-1369. 1991.

453. Kinzler. K. W., Nilbert, M. C., Su, L-K., Vogelstein, B., Bryan. T. M.,Levy, D. B., Smith, K. J.. Preisinger, A. C., Hedge, P., McKechnie, D.,Finnibar, R., Markham, A., Groffen, J., Boguski. M. S., Altschul, S. F.,Horii, A., Ando, H.. Miyoshi, Y., Miki, Y., Nishisho, I., and Nakamura, Y.Identification of FAP locus genes from chromosome 5q21. Science (Washington DC), 255: 661-665, 1991.

454. Nishisho, I., Nakamura, Y., Miyoshi, Y., Miki, Y.. Ando, H., Horii, A.,Koyama, K., Ursunomiya, J.. Baba, S., Hedge, P.. Markham, A., Krush, A.J., Petersen, G., Hamilton. S. R., Nilbert, M. C, Levy, D. B., Bryan, T. M.,Preisinger, A. C, Smith, K. J., Su, L-K., Kinzler, K. W. and Vogelstein, B.Mutations of chromosome 5q21 genes in FAP and colorectal cancer patients. Science (Washington, DC), 253: 665-669, 1991.

455. Groden, J., Thilveria, A., Samowitz, W., Carlson. M., Gilbert, L., Albertsen,H., Joslyn, G., Stevens, J., Spinn. L., Robertson, M., Sargeant, L., Krapcho,K., Wolff, E., Buri. R., Hughes, J. P., Warrington, J., McPherson, J.,Wasmuth, J., Le Pasller, D., Abderrahim, H., Cohen, D.. Leppert, M., andWhite, R. Identification and charcterization of the familial adenomatouspolyposis coli gene. Cell, 55: 1-20, 1991.

456. LaForgia, S., Morse, B., Levy, J., Barnea, G., Cannizzaro, L. A., Li, F.,Nowell, P. C.. Boghosian-Sell, L., Click, J., Weston, A., Harris, C. C,Drabkin, H., Patterson, D., Croce. C. M., Schlessinger, J., and Huebner, K.Receptor-linked protein-tyrosine-phosphatase, PTPy, is a candidate tumorsuppressor at human chromosome region 3p21. Proc. Nati. Acad. Sci. USA,88: 5036-5040, 1991.

457. Reynolds, S. H., Stowers, S. J., Patterson, R. M.. Maronpot, R. R.,Aaronson, S. A., and Anderson, M. W. Activated oncogenes in B6C3F,mouse liver tumors: implications for risk assessment. Science (WashingtonDC), 2J7: 1309-1316, 1987.

458. Fox, T. R., Schumann, A. M., Watanabe, P. G., Yano. B. L., Mäher,V.M., and McCormick, J. J. Mutational analysis of the H-ras oncogene inspontaneous C57BL/6 X C3H/He mouse liver tumors and tumors inducedwith genotoxic and nongenotoxic hepatocarcinogens. Cancer Res., 50:4014-4019, 1990.

459. You, M., Candrian, U., Maronpot, R. R., Stoner, G. D., and Anderson, M.W. Activation of the Ki-ras protooncogene in spontaneously occurring andchemically induced lung tumors of the strain A mouse. Proc. Nati. Acad.Sci. USA, 86: 3070-3074, 1989.

460. Belinsky, S. A., Devereux, T. R., Maronpot, R. R., Stoner, G. D., andAnderson, M. W. Relationship between the formation of promutagenicadducts and the activation of the K-nu protooncogene in lung tumors fromA/J mice treated with nitrosamines. Cancer Res., 49: 5305-5311, 1989.

461. Devereux. T. R., Anderson, M. W.. and Belinsky, S. A. Role of ras protooncogene activation in the formation of spontaneous and nitrosamine-induced lung tumors in the resistant C3H mouse. Carcinogenesis (Lond.),in press, 1991.

462. Sukumar, S., Perantoni, A., Reed, C., Rice, J. M., and Wenk, M. L.Activated K-ros and N-rai oncogenes in primary renal mesenchymal tumorsinduced in F344 rats by methyl(methoxymethyl)nitrosamine. Mol. CellBiol., 6:2716-2720, 1986.

463. Stowers, S. J., Glover, P. L., Reynolds, S. H., Boone, L. R., Maronpot, R.R., and Anderson, M. W. Activation of the K-raj oncogene in rat esophagealpapillomas induced by methylbenzylnitrosamine. Cancer Res., 47: 3212-3219, 1987.

464. Dandekar, S., Sukumar. S.. Zarbl. H., Young. L. J., and Cardiff, R. D.Specific activation of the cellular Harvey-ras oncogene in dimethylbenzan-thracene-induced mouse mammary tumors. Mol. Cell Biol., 6: 4104-4108,1986.

465. Reynolds, S. H., Patterson, R. M., Mennear, J. H.. Maronpot, R. R., andAnderson, M. W. ras gene activation in rat tumors induced by benzidinecongeners and derived dyes. Cancer Res., 50: 266-272, 1990.

466. Bonham, K., Embry, T., Gibson, D., Jaffe, D. R., Roberts, R. A., Cress, A.E., and Bowden, G. T. Activation of the cellular Harvey ras gene in mouseskin tumors initiated with urethane. Mol. Carcinog., 2: 34-39, 1989.

467. Wang, Y., You. M., Reynolds, S. H., Stoner, G. D., and Anderson, M. W.Mutational activation of the cellular Harvey ras oncogene in rat esophagealpapillomas induced by methylbenzylnitrosamine. Cancer Res.. 50: 1591-1595, 1990.

468. Muir, C. S. Changing international patterns of cancer incidence. In: J. G.Fortner and J. E. Rhoads (eds.). Accomplishments in Cancer Research 1988Prize Year. General Motors Cancer Research Foundation, pp. 126-144.Philadelphia: J. B. Lippincott Co., 1988.

5044s

on April 2, 2020. © 1991 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Page 23: Chemical and Physical Carcinogenesis: Advances …...CARCINOGENESIS Table 1 Examples of advances in chemical and physical carcinogenesis" A. Cancer etiology I. Life-style factors (tobacco

1991;51:5023s-5044s. Cancer Res   Curtis C. Harris  Perspectives for the 1990sChemical and Physical Carcinogenesis: Advances and

  Updated version

  http://cancerres.aacrjournals.org/content/51/18_Supplement/5023s

Access the most recent version of this article at:

   

   

   

  E-mail alerts related to this article or journal.Sign up to receive free email-alerts

  Subscriptions

Reprints and

  [email protected] at

To order reprints of this article or to subscribe to the journal, contact the AACR Publications

  Permissions

  Rightslink site. Click on "Request Permissions" which will take you to the Copyright Clearance Center's (CCC)

.http://cancerres.aacrjournals.org/content/51/18_Supplement/5023sTo request permission to re-use all or part of this article, use this link

on April 2, 2020. © 1991 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from