The Health Consequences of Using Smokeless Tobacco A Report of the Advisory Committee to the Surgeon General 1986 U.S. DEPARTMENT OF HEALTH AND HUMAN SERVICES Public Health Service Bethesda, Maryland 20992 NIH Publication No. 96-2874 April 1996 CONTENTS Foreword ......................................... ..v Preface ........................................... . Acknowledgments ................................. ..ix Introduction, Overview, and Conclusions ................ xvii Chapter 1. Prevalence and lbnds of Smokeless lbbaccoUsei.ntheUnitedStates . . . . .l Chapter 2. Carcinogenesis Associated With Smokeless Tobacco Use . . . . .29 Chapter 3. Noncancerous and Precancerous Oral Health Effects Associated With Smokeless Tobacco Use . . . . . . . .95 Chapter 4. Nicotine Exposure: Pharmacokinetics, Addiction, and Other Physiologic Effects . . ,137 Index..................... . . . . . . . . . . . . . ...187 . . . 111 FOREWORD This report on The Health Consequences of Using Smokeless lbbamo completes the Public Health Service's initial examination of smokeless tobacco's role in the causation of cancer, noncancerous and precancer- ous oral diseases or conditions, addiction, and other adverse health effects. Almost 30 years after the Public Health Service's first state ment on the health effects of cigarette smoking, it is now possible to issue the first comprehensive, indepth review of the relationship between smokeless tobacco use and health. Ironically, while cigarette smoking has declined during the past 20 years, the production and apparent consumption of smokeless tobacco products have risen significantly. These increases are in marked con- trast to the decline in smokeless tobacco use in the United States during the first half of this century. Indeed, smokeless tobacco products, par- ticularly chewing tobacco and snuff, have recently emerged as popular products for the first time since the turn of the century. National esti- mates indicate that at least 12 million Americans used some form of smokeless tobacco during 1985 with use increasing especially among male adolescents and young male adults. The increased use and appeal of this product assume major public health significance because the evidence reveals that smokeless tobacco can cause oral cancer, can lead to the development of oral leukoplakias and other oral conditions, and can cause addiction to nicotine. The strength of the association between these conditions and smokeless tobacco use combined with the upward trend in this behavior incites the same alarm as was true with the knowledge that spitting spread tuber- culosis. That concern led to the original public rejection of tobacco chewing and dipping as unsanitary and antisocial. It is critical that our society prevent the use of this health hazard and avoid the tragic mistake of replacing the ashtray with the spittoon. This report is the work of numerous experts within the Department of Health and Human Services and in the non-Federal scientific com- munity. I express my gratitude for their contributions. C. Everett Koop, M.D. U.S. Surgeon General PREFACE This report discusses the health consequences of smokeless tobacco use. It constitutes a comprehensive review by an Advisory Committee to the Surgeon General of the available scientific literature to determine whether using smokeless tobacco increases the risk of cancer and non- cancerous oral diseases and effects, leads to addiction and dependence, and contributes to other health consequences. AFTER A CAREFUL EXAMINATION OF THE RELEVANT EPIDEMIOLOGIC, EXPERIMENTAL, AND CLINICAL DATA, THE COMMITTEE CONCLUDES THAT THE ORAL USE OF SMOKELESS TOBACCO REPRESENTS A SIGNIFICANT HEALTH RISK. IT IS NOT A SAFE SUBSTITUTE FOR SMOK- ING CIGARETTES. IT CAN CAUSE CANCER AND A NUMBER OF NONCANCEROUS ORAL CONDITIONS AND CAN LEAD TO NICOTINE ADDICTION AND DEPENDENCE. The major overall conclusions of this report are the following: 1. It is estimated that smokeless tobacco was used by at least 12 million people in the United States in 1985 and that half of these were regular users. The use of smokeless tobacco, particularly moist snuff, is increasing, especially among male adolescents and young male adults. 2. The scientific evidence is strong that the use of snuff can cause cancer in humans. The evidence for causality is strongest for cancer of the oral cavity, wherein cancer may occur several times more frequently in snuff dippers compared to nontobacco users. The excess risk of cancer of the cheek and gum may reach nearly fiftyfold among long-term snuff users. 3. Some investigations suggest that the use of chewing tobacco may also increase the risk of oral cancer, but the evidence is not so strong and the risks have yet to be quantified. 4. Experimental investigations reveal potent carcinogens in smoke less tobacco. These include nitrosamines, polycyclic aromatic hydrocarbons, and radiation-emitting polonium. The tobacco- specific nitrosamines often have been detected at levels 100 or more times higher than Government-regulated levels of other nitrosamines permitted in foods eaten by Americans. vii 5. Smokeless tobacco use can lead to the development of oral leuko- plakias (white patches or plaques of the oral mucosa), particularly at the site of tobacco placement. Based on evidence from several studies, a portion of leukoplakias can undergo transformation to dysplasia and further to cancer. 6. Gingival recession is a commonly reported outcome of smokeless tobacco use. 7. A number of studies have shown that nicotine exposure from smoking cigarettes can cause addiction in humans. In this regard, nicotine is similar to other addictive drugs such as morphine and cocaine. Since nicotine levels in the body resulting from smokeless tobacco use are similar in magnitude to nicotine levels from cigarette smoking, it is concluded that smokeless tobacco use also can be addictive. Besides, recent studies have shown that nicotine administered orally has the potential to produce a physiologic dependence. 8. Some evidence suggests that nicotine may play a contributory or supportive role in the pathogenesis of coronary artery and periph- eral vascular disease, hypertension, peptic ulcers, and fetal mortal- ity and morbidity. . . . Vlll ACKNOWLEDGMENTS This report was prepared by the Department of Health and Human Services under the direction and general editorship of Joseph W. Cullen, Ph.D., Chairman of the Advisory Committee on the Health Conse quences of Using Smokeless lbbacco. The following individuals were members of the Advisory Committee: Catherine S. Bell, M.S., Acting Chief, Prevention Research Branch, Division of Clinical Research, National Institute on Drug Abuse, Rockville, Maryland William Blot, Ph.D., Chief, Biostatistics Branch, Division of Cancer Etiology, National Cancer Institute, Bethesda, Maryland James P. Carlos, D.D.S., M.P.H., Associate Director, Epidemiology and Oral Disease Prevention Program, National Institute of Dental Research, Bethesda, Maryland Joseph W. Cullen, Ph.D., Chairman of the Advisory Committee, Dep- uty Director, Division of Cancer Prevention and Control, National Cancer Institute, Bethesda, Maryland J. David Erickson, D.D.S., M.P.H., Ph.D., Chief, Birth Defects and Genetic Diseases Branch, Centers for Disease Control, Atlanta, Georgia Manning Feinleib, M.D., Dr.P.H., Director, National Center for Health Statistics, Hya ttsville, Maryland Jerome Jaffe, M.D., Director, Addiction Research Center, National Institute on Drug Abuse, Baltimore, Maryland Michael McGinnis, M.D., M. A., M.P.P., Deputy Assistant Secretary for Health, Director, Office of Disease Prevention and Health Promo tion, United States Public Health Service, Washington, D.C. Robert Mecklenburg, D.D.S., M.P.H., Assistant Surgeon General, Chief Dental Officer, United States Public Health Service, Rockville, Maryland Susan Sieber, Ph.D., Deputy Director, Division of Cancer Etiology, National Cancer Institute, Bethesda, Maryland Sumner J. Yaffe, M.D., Director, Center for Research for Mothers and Children, National Institute of Child Health and Human Develop- ment, Bethesda, Maryland The Advisory Committee was supported by the following technical experts in the areas listed below: ix Carcinogenesis William Blot, Ph.D. (Coordinator), Chief, Biostatistics Branch, Division of Cancer Etiology, National Cancer Institute, Bethesda, Maryland Gayle Boyd, Ph.D., Research Psychologist, Division of Cancer Preven- tion and Control, National Cancer Institute, Silver Spring, Maryland Virginia L. Em&r, Ph.D., Associate Professor of Epidemiology, Uni- versity of California, San Francisco, California Curtis Harris, M.D., Chief, Laboratory of Human Carcinogenesis, Divi- sion of Cancer Etiology, National Cancer Institute, Bethesda, Maryland Dietrich Hoffmann, Ph.D., Associate Director, Chemistry and Bio- chemistry, Naylor Dana Institute for Disease Prevention, American Health Foundation, Valhalla, New York Kenneth Rothman, Dr.P.H., Professor of Family and Community Medicine, University of Massachusetts Medical School, Worcester, Massachusetts David Schottenfeld, M.D., Chief, Division of Epidemiology, Memorial Sloan-Kettering Cancer Center, New York, New York Steven R. Tmnenbaum, Ph.D., Professor of Toxicology and Food Chemistry, Department of Applied Biological Sciences, Massachu- setts Institute of Technology, Cambridge, Massachusetts David B. Thomas, M.D., Dr. PH., Head Program in Epidemiology, Fred Hutchinson Cancer Research Center, Seattle, Washington Non-Cancer Od Effects Arden G. Christen, D.D.S., M.S.D., M.A., Chairman, Department of Preventive Dentistry, Indiana University Dental School, Indianapo lis, Indiana Stephen B. Corbin, D.D.S., M.P.H., Chief, Dental Disease Prevention Activity, Center for Preventive Services, Centers for Disease Control, Rockville, Maryland Robert 0. Greer, Jr., D.D.S., Sc.D., Professor, Division of Oral Pathol- ogy and Oncology, University of Colorado Health Sciences Center, Denver, Colorado Lireka P. Joseph, Dr.P.H., Medical Radiation Specialist, Food and Drug Administration, Rockvihe, Maryland Stephen S. Kegeles, Ph.D., Professor, Department of Behavioral Sci- ences and Community Health, University of Connecticut Health Center, Farmington, Connecticut Matthew Kinnard, Ph.D., Health Science Administrator, Periodontal and Soft Tissue Diseases Research Branch, National Institute of Dental Research, Bethesda, Maryland Dushanka Kleinman, D.D.S., M.Sc.D., Special Assistant to the Asso ciate Director for Program Coordination, Epidemiology and Oral Disease Prevention Program, National Institute of Dental Research, Bethesda, Maryland X Robert Mecklenburg, D.D.S., M.P.H. (Coordinator), Assistant Surgeon General, Chief Dental Officer, United States Public Health Service, Rmkdle, Maryland Kathleen L. Schroeder, D.D.S., M.S., Assistant Professor, Department of Oral Biology, Ohio State University College of Dentistry, Columbus, Ohio Addiction and Dependence Catherine S. Bell, M.S. (Coordinator), Acting Chief, Prevention Re search Branch Division of Clinical Research, National Institute on Drug Abuse, RockviRe, Maryland Neal Benowitz, M.D., Associate Professor of Medicine, Chief of Clinical Pharmacology and Experimental Therapeutics, University of Cali- fornia, San Francisco, california Anthony Biglan, Ph.D., Research Scientist, Oregon Research Institute, Eugene, Oregon John Grabowski, Ph.D., Associate Professor, Department of Psychiatry, School of Medicine, Louisiana State University, Shreveport, Louisiana Steven Gust, Ph.D., Reseaxh Associate, Department of Psychiatry, University of Minnesota, Minneapolis, Minnesota Jack Hennmgfield, PhD., Chief, Biology of Dependence and Abuse Potential Assessment Laboratory, Addiction Research Center, National Institute on Drug Abuse, Baltimore, Maryland Lynn Kozlowski, PhD., Senior Scientist, The Smoking Research Pro gram, Addiction Reseamh Foundation, Toronto, Ontario, Canada Norman A. Krasnegor, PhD., Chief, Human Learning and Behavioral Branch, Center for Research for Mothers and Children, National Institute for Child Health and Human Development, Bethesda, Maryland Ovide Pomerleau, PhD., Chief of Psychology Service, Veterans Admin- istration Medical Center, Newington, Connecticut The following individuals prepared draft chapters or portions of the report as indicated: Chapter I. Prevalence and Fends of Smokeless lbbacco Use in the United States Gayle Boyd, PhD., Research Psychologist, Division of Cancer Preven- tion and Control, National Cancer Institute, Silver Spring, Maryland Rebecca Cline, PhD., Senior Associate, Prospect Associates, Rockille, Maryland Charles Darby, M.A., `Ikhnical`Support Staff Coordinator for Addic- tion and Dependence Experts, Senior Associate, Prospect Associates, RmkvilIe, Maryland Margaret Mattson, Ph.D., Special Assistant for Science, Office of the Director, Division of Cancer Prevention and Control, National Cancer Institute, Bethesda, Maryland Chapter II. Carcirwgenesis Associated With Smokeless Tbbacco Use William Blot, Ph.D., Chief, Biostatistics Branch, Division of Cancer Etiology, National Cancer Institute, Bethesda, Maryland Virginia L. Ems&, Ph.D., Associate Professor of Epidemiology, University of California, San Francisco, California Curtis Harris, M.D., Chief, Laboratory of Human Carcinogenesis, Division of Cancer Etiology, National Cancer Institute, Bethesda, Maryland Stephen S. Hecht, Ph.D., Division of Chemical Carcinogenesis, Naylor Dana Institute for Disease Prevention, American Health Foundation, Valhalla, New York Dietrich Hoffmann, Ph.D., Associate Director, Chemistry and Bio chemistry, Naylor Dana Institute for Disease Prevention, American Health Foundation, Valhalla, New York Kenneth Rothman, Dr.P.H., Professor of Family and Community Medi- cine, University of Massachusetts Medical School, Worcester, Massachusetts David Schottenfeld, M.D., Chief, Division of Epidemiology, Memorial Sloan-Kettering Cancer Center, New York, New York Steven R. Xannenbaum, PhD., Professor of Tbxicology and Food Chemistry, Department of Applied Biological Sciences, Massachu- setts Institute of %chnology, Cambridge, Massachusetts David B. Thomas, M.D., Dr.P.H., Head, Program in Epidemiology, Fred Hutchinson Cancer Research Center, Seattle, Washington Deborah M. Winn, Ph.D., Epidemiologist, Survey Planning and Devel- opment Branch National Center for Health Statistics, Hyattsville, Maryland Chapter III. Noncancerous and Precancerous Oral Health Effects Associated With Smokeless llbbacco Use Stephen B. Corbin, D.D.S., M.P.H., Chief, Dental Disease Prevention Activity, Center for Preventive Services, Centers for Disease Control, RmkvilIe, Maryland Lireka I? Joseph, Dr.P.H., Medical Radiation Specialist, Food and Drug Administration, Rockvihe, Maryland Dushanka Kleimnan, D.D.S., M.Sc.D., Special Assistant to the Asso ciate Director for Program Coordination, Epidemiology and Oral Disease Prevention Program, National Institute of Dental Research, Bethesda, Maryland Chapter IV. Nicotine Exposure: Pharmacokinetics, Addiction, and Other Physiologic Effects Neal Benowitz, M.D., Associate Professor of Medicine, Chief of Clinical Pharmacology and Experimental Therapeutics, University of Cali- fornia, San Francisco, California Anthony Biglan, PhD., Research Scientist, Oregon Research Institute, Eugene, Oregon xii Jack Henningfield, Ph.D., Chief, Biology of Dependence and Abuse Potential Assessment Laboratory, Addiction Research Center, National Institute on Drug Abuse, Baltimore, Maryland Margaret Mattson, PhD., Special Assistant for Science, Office of the Director, Division of Cancer Prevention and Control, National Cancer Institute, Bethesda, Maryland The Advisory Committee and authors acknowledge with gratitude the following distinguished scientists, physicians, and others who lent their support in the development of this report by contributing critical reviews of the manuscript or assisting in other ways: John Bailar, M.D., Ph.D., Lecturer, Department of Biostatistics, Har- vard School of Public Health, Boston, Massachusetts Enriqueta C. Bond, PhD., Director, Division of Health Promotion and Disease Prevention, Institute of Medicine, Washington, D.C. Lester Breslow, M.D., M.P.H., Professor Emeritus, School of Public Health, University of California, Los Angeles, California David M. Burns, M.D., Associate Professor of Medicine, Division of Pulmonary and Critical Care Medicine, University of California, San Diego Medical Center, San Diego, California Greg Connolly, D.M.D., M.P.H., Director, Division of Dental Health, Massachusetts Department of Public Health Boston Massachusetts D. Layten Davis, PhD., Director, University of Kentucky, Ylbbacco and Health Research Institute, Lexington, Kentucky Miriam Davis, PhD., Program Analyst, Office of Health Planning and Evaluation, Office of the Assistant Secretary for Health, Washing- ton, D.C. Vincent T. DeVita, Jr., M.D., Director, National Cancer Institute, Bethesda, Maryland Joseph F. Fraumeni, Jr., M.D., Associate Director for Epidemiology and Biostatistics, Division of Cancer Etiology, National Cancer Insti- tute, Bethesda, Maryland Thomas J. Glynn, PhD., Program Director for Smoking Research, Division of Cancer Prevention and Control, National Cancer Insti- tute, Bethesda, Maryland Peter Greenwald, M.D., Dr.P.H., Director, Division of Cancer Preven- tion and Control, National Cancer Institute, Bethesda, Maryland Ellen R. Grits, PhD., Director, Division of Cancer Control, UCLA Jonsson Comprehensive Cancer Center, Los Angeles, Cahfornia Dianne G. Lindewall, Ph.D., Program Analyst, Office of Health Plan- ning and Evaluation, Office of the Assistant Secretary for Health, Washington, D.C. Marilyn M. Massey, M.P.H., `Ikchnical Support Staff Coordinator for the Advisory Committee, Senior Associate, Prospect Associates, lbdwille, Maryland . . . XlJl Gardner C. McMillan, M.D., Associate Director for Etiology, Arterio sclerosis, and Hypertension, National Heart, Lung, and Blood Insti- tute, Bethesda, Maryland Jens J. Pindborg, D.D.S., Dr. Odont., Professor of Oral Pathology, Royal Dental College, Copenhagen Panum Institute, Copenhagen Denmark John M. Pinney, Executive Director, Institute for the Study of Smoking Behavior and Policy, John F. Kennedy School of Government, Har- vard University, Cambridge, Massachusetts Earl S. Polk&, Sc.D., President, Pollack Associates Ltd., Bethesda, Maryland Richard J. Riseberg, Chief Counsel United States Public Health Ser- vice, Rochille, Maryland Thomas L. Robertson, M.D., Chief, Cardiac Diseases Branch, National Heart, Lung, and Blood Institute, Bethesda, Maryland Beatrice A. Rouse, PhD., Epidemiologist, Division of Epidemiology and Statistical Analysis, National Institute on Drug Abuse, Rock- vilIe, Maryland Donald R. Shopland, Acting Director, Office on Smoking and Health, United States Public Health Service, Rockvile, Maryland Sol Silverman Jr., M.A., D.D.S., Professor and Chairman of Oral Medi- cine, University of California, San Francisco, California Chester Slaughter, M.A., Deputy Regional Inspector General for Analy- sis and Inspections, Office of the Inspector General DaIlas, Texas K. W. Stephen, D.D.Sc., H.D.D.R.C.P.S., Head of Oral Medicine and Pathology, University of Glasgow, Glasgow, Scotland Ronald W. Wilson, M.A., Director, Division of Epidemiology and Health Promotion, National Center for Health Statistics, Hyattsville, Maryland Ernst L. Wynder, M.D., President, American Health Foundation, New York, New York Mar& Zitans, M.&z., Program Analyst, Office of the Inspector General Washington, D.C. The Advisory Committee and authors also acknowledge several organizations for their contributions in identifying relevant literature and/or in offering critical reviews of the manuscript. Institute of Medicine, National Academy of Sciences, for providing through its Board on Health Promotion and Disease Prevention a cri- tique of the Advisory Committee's review methods and drafts of the report. International Agency for Research on Cancer (IARC), Lyon, France, for providing IARC Monograph No. 37 on the Evaluation of the Carcino genie Risk of Chemicals to Humans: Tobacco Habits Other Than Smoking Betel-Quid and Areca-Nut Chewing and Some Related Nitrosammes, September 1985. National Center for Health Statistics and National Institute on Drug Abuse, for providing data on the prevalence and trends of smokeless tobacco use in the United States. Office on Smoking and Health, for bibliographic citations and other documents pertinent to the literature review. Smokeless Tobacco Council, Inc. (Michael J. Kerrigan, President), for providing approximately 15,000 citations considered relevant by a number of the smokeless tobacco companies, including the collection of testimonies representing the smokeless tobacco industry's position concerning the health consequences of smokeless tobacco. The Advisory Committee and authors also acknowledge the contribu- tions of the following staff members and others who assisted in the preparation of the report: Kathy Bauman, Editorial Assistant to the Deputy Director, Division of Cancer Prevention and Control, National Cancer Institute, Bethesda, Maryland Julie Begtm, Research Assistant, Prospect Associates, Rockville, Maryland Barbara Clark, M.S., M.P.H., Technical Support Staff Coordinator for Non-Cancer Oral Effects Experts, Prospect Associates, Rockvihe, Maryland Shelley Clark, Conference Coordinator, Prospect Associates, Rockvihe, Maryland Nancy Cowan Conference Coordinator, Prospect Associates, Rockvihe, Maryland Kathleen Edmunds, Library Besearch Assistant, Prospect Associates, RockviUe, Maryland Robyn Ertwine, Librarian-Information Specialist, Prospect Associates, RockviNe, Maryland Catherine Fisher, Secretary, Prospect Associates, Rockvihe, Maryland Barbara Hayden, Secretary, Prospect Associates, Rockvihe, Maryland Douglas Hunter, Library Assistant, Prospect Associates, Kockvihe, Maryland Karen Jacob, Editor, Prospect Associates, Rockvihe, Maryland Margaret Johnson, `Typist, Prospect Associates, Rockville, Maryland Mary Johnson, Typist, Prospect Associates, Rockvihe, Maryland Virginia Knill, Typist, Prospect Associates, RockviIle, Maryland Solomon Levy, M.P.H., Associate Director, Office of the Surgeon General, Rockvihe, Maryland Paula Margus, Library Research Assistant, Prospect Associates, Rock- ville, Maryland Nancy McCormick-Picket& M.A., %&nicsl Support Staff Coordinator for Carcinogenesis Experts, Prospect Associates, Rockvihe, Maryland Lee McPherson, Typist, Prospect Associates, Rockvihe, Maryland xv Robbie Morsel&, Library Assistant, Prospect Associates, Rockville, Maryland Elizabeth Mugge, Secretary to the Deputy Director of the Division of Cancer Prevention and Control, National Cancer Institute, Bethesda, Maryland Arthur Nelson, Library Research Assistant, Prospect Associates, Fhkville, Maryland Ro NemethCoslett, Ph.D., Visiting Scientist, Biology of Dependence and Abuse Potential Assessment Laboratory, Addiction Research Center, National Institute on Drug Abuse, Baltimore, Maryland Simon Plog, l)pist, Prospect Associates, Rockvihe, Maryland Nancy Rudner, M.S.N., M.P.H., 7hchnicaI Support Staff Assistant, Prospect Associates, Rockvihe, Maryland Estelle Schwalb, `Ilxhnical Writer-Editor, Prospect Associates, Rock- ville, Maryland Barbara Shapiro, Editor, Prospect Associates, Rockville, Maryland John Shea, Ph.D., Technical Writer-Editor, Prospect Associates, Rock- ville, Maryland Dorritt Silber, Secretary, Prospect Associates, Rockvihe, Maryland Dori Steele, Proofreader, Prospect Associates, Rockville, Maryland Deborah S. Swansburg, Program Officer, Institute of Medicine, National Academy of Sciences, Washington, D.C. Xvi INTFlODlJC'TlON, OVERVIEW, AND CONCLUSIONS DEVELOPMENT AND ORGANIZATION OF THE REPORT This report from the Surgeon General's Advisory Committee on the Health Consequences of Using Smokeless Tobacco represents the first comprehensive assessment of the biomedical and behavioral literature describing experimental and human evidence on the health conse quences of using smokeless tobacco. The content of this report is the work of numerous experts within the Department of Health and Human Services as well as distinguished scientists outside the organization. Each chapter of the report was prepared based on manuscripts writ- ten by scientists who are recognized for their understanding of the spe cific content areas. Manuscripts were subjected to extensive peer review by a large number of experts in the specific areas of interest. The report includes a "Preface" that presents the essence of the entire report and an "Introduction, Overview, and Conclusions." The body of the report consists of the following four chapters: o Chapter l-Prevalence and Trends of Smokeless Tobacco Use in the United States o Chapter 2-Carcinogenesis Associated With Smokeless ?bbacco Use o Chapter 3-Noncancerous and Precancerous Oral Health Effects Associated With Smokeless Tobacco Use o Chapter 4-Nicotine Exposure: Pharmacokinetics, Addiction, and Other Physiologic Effects HJSTORICAL PERSPECTIVE The use of smokeless tobacco is a worldwide practice with numerous variations in the nature of the product used as well as in the customs associated with its use. In the United States, smokeless tobacco con- sists of chewing tobacco and snuff. The predo minant mode of use of these nonsmoked tobaccos is oral, although they may be placed in or inhaled into the nasal cavity. Tobacco sniffing, however, has been and remains a rare practice in the United States. Xvii Smokeless tobacco was used in the United States in the early 1600's when snuff made its way to the Jamestown Colony in Virginia through the efforts of John RoIfe in 1611(l). Evidence of tobacco chewing, how- ever, was not found until a century later in 1704 (2). The use of tobacco, including smokeless tobacco, has been controver- sial since its introduction. In the past, tobacco use was considered by some as beneficial As early as 3500 B.C., there are indications that tobacco was an article of established value to the inhabitants of Mexico and Peru. It appears that people who frequently Iacked sufficient food alleviated their hunger pains by chewing tobacco (3). Smokeless tobacco was also thought to have several medicinal uses. Among Native Ameri- cans, for example, chewing tobacco was used to alleviate toothaches, disinfect cuts, and relieve the effects of snake, spider, and insect bites (4). Moreover, during the 19th and early 20th centuries in America, dental snuff was advertised to relieve toothache pain; to cure neuralgia, bleed- ing gums, and scurvy; and to preserve and whiten teeth and prevent decay (1). On the other hand, tobacco use historically has had numerous adver- saries, inchiding the following (1): o In 1590 in Japan tobacco was prohibited. Users lost their property and were jaiIed o King James VI of Scotland in the early 1600's was a strong anti- smoking advocate who increased taxes on tobacco 4,000 percent in an attempt to reduce the quantity imported to England. o In 1633, the Sultan Murad IV of Turkey made any use of tobacco a capital offense, punishable by death from hanging, beheading, or starvation. He maintained that tobacco caused infertility and reduced the fighting capabilities of his soldiers. o The Russian Czar Michael Fedorovich, the first Romanov (1613-1645). prohibited the sale of tobacco, stating that users would be subjected to physical punishment and that persistent users would be killed. o A Chinese law in 1683 threatened that anyone possessing tobacco would be beheaded. o During the mid-1600's, Pope Urban VIII banned the use of snuff in churches, and Pope Innocent X attacked its use by priests in the Catholic Church. o Other religious groups also banned snuff use: John Wesley, the founder of Methodism, attacked its use in Ireland; the Mormons, Seventh-Day Adventists, Parsees and Sikhs of India, Buddhist monks of Korea, members of the Isai Li sect of China, and some Ethiopian Christian sects forbade the use of tobacco. . . . XVIU o Frederick the Great, King of Prussia, prevented his mother, the Dowager Queen of Prussia, from using snuff at his coronation in 1790. o Louis XV, ruler of France from 1723 to 1774, banned snuff use from the Court of France. Scientific observations concerning the health effects of smokeless tobacco use were first noted in 1761 by John Hill, a London physician and botanist who reported five cases of polypuses, a "swelling in the nostril that was hard, black and adherent with the symptoms of an open cancer" (5). He concluded that nasal cancer could develop as a conse quence of tobacco snuff use (sniffing). Evidence that suggested a possible association between smokeless tobacco use and oral conditions in North America and Europe was not reported until 1915 when Abbe identified several tobacco chewers among a series of oral cancer patients and commented that smokeless tobacco use may be a risk factor for this cancer (6). In the late 1930's. Ahblom observed in Sweden that more patients with buccal, gingival, and `mandibular" cancers than with other cancers reported the use of snuff or chewing tobacco (?L In the United States, case reports of oral cancer among users of snuff or chewing tobacco appeared in the early 1940's (8). The first epidemiologic study of smokeless tobacco was not conducted until the early 1950's (9). Since that time, several scientists have described a pattern of increased risk of oral cancer among smoke less tobacco users. Investigations of other possible health effects of smokeless tobacco use (e.g., noncancerous oral effects, addiction, and other physiologic consequences) are more recent subjects of scientific inquiry that have been undertaken primarily in the past two decades. A brief review of the health consequences of smokeless tobacco was pre sented in the 1979 Surgeon General's report on smoking and health (10). Since that review, the results of additional studies addressing the role of smokeless tobacco in health have become available and thus provide the basis of this current comprehensive review. REVIEW METHODS For the purpose of evaluating the scientific evidence to be included in this report, the Advisory Committee called upon the same criteria to determine causality as have been used for a number of Surgeon General's reports on smoking for the past two decades. The following criteria were used as the primary guidelines for assessing whether any associations between smokeless tobacco use and each of the disease areas or health conditions under e xamination were likely to be causal in Mtwe: xix o Consistency of the association-similar observations by multiple investigators in different locations and situations, at different times, and using different methods of study. o Strength of the association-high ratio of disease rate for thepopu- lation exposed to the suspected risk factor compared to the popula- tion unexposed to the risk factor. o Specificity of the association-associations with the exposure exist for a specific or limited set of diseases, and associations with the disease exist for a specific or limited set of exposures. o Temporal relationship of the association-exposure to the suspected etiologic factor precedes the disease. o Coherence of the association-epidemiologic observations are con- sonant with all else that is known about the disease. In addition to these criteria, the general principles employed by the International Agency for Resear& on Cancer (IARC)* in evaluating the carcinogenic risk of chemicals or complex mixtures (table 1) were used as needed to supplement the primary causation criteria (11). The use of smokeless tobacco products in the United States was wide spread until the end of the 19th century. With the advent of antispitting laws, loss of social acceptability, and increased popularity of cigarette smoking, its use declined rapidly in this century. However, recent na- tional data indicate a resurgence in smokeless tobacco habits with more than 12 million persons estimated as users of some form of smokeless tobacco in 1985. An upward trend in use is emerging, particularly among young males. Given the evidence that smokeless tobacco is regaining popularity, serious questions have been raised about its adverse health effects. Most notably, this behavior has been linked to cancer, specifically, oral cancer. Analytic epidemiologic studies now indicate that the use of oral snuff increases the risk of oral cancer several fold and that among long- term snuff dippers the excess risk of cancers of the cheek and gum may reach nearly fiftyfold. This conclusion is consistent with the judgment of a recent working group of the IARC, which assessed the carcinogenic risk associated with tobacco habits other than smoking (11). The conclusion that smokeless tobacco causes cancer results from several lines of evidence: the presence of high levels of carcinogens in smokeless tobacco; the metabolic conversion of products of smokeless * The IARC was established in 1965 b within the framework of the World Hea i the World Health Assembly as an inde th Organization. It conducts a program o P K ndentl financed organization researc concentrating particu- larly on the epidemiology of cancer and the study of potential carcinogens in the human environment. xx TABLE L-General F'rinciples in Evaluting Carcinogenic Risk of Chemicals or Complex Mixture8 (Intemational Agency for R.4wal& on Cancer) o Evidence for carcinogenicity in experimental animals: - Qualitative aspects: (a) Experimental parameters under which chemical was tested. (b)Consistency with which chemical shown to be ?????*???*??? (12) Spectrum of neoplastic response. (d) Stage of tumor formation in which chemical involved. (e) Role of modifying factors. - Hormonal carcinogenesis. - Complex mixtures. - Quantitative aspects; increasing incidence of neoplasms with increasing exposure. o Evidence for activity in short-term tests: - Use of valid test system. - Sufficiently wide dose range and duration of exposure to the agent and appropriate metabolic system employed in test. - Use of appropriate controls. - Specification of the purity of the compound, and in the case of complex mixtures, source and representativeness of sample tested. o Evidence of carcinogenicity in humans: - For studies showing positive association: (a) Existence of no identifiable bias. (b) Possibility of positive confounding considered. (c) Association unlikely to be due to chance alone. (d) Association is strong. (e) Existence of dose-response relationship. - For studies showing no association: (a) Existence of no identifiable negative bias. (b) Possibility of negative wnfounding considered. (c) Possible effects of misclassification of exposure or outcome have been ???????? tobacco into genotoxic agents; the consistency of the oral cancer- smokeless tobacco association across epidemiologic investigations con- ducted in diverse locations; the trend in increasiq oral cancer risk with duration of exposure; the strength of the association with oral cancer and the occurrence of the highest risks for cancers at the anatomic sites where the tobacco expoams are the greatest. In addition, a number of clinical observations and studies show an association between smokeless tobacco use and some noncancerous and precancerous oral health conditions. The development of a portion of oral leukoplakias in both teenage and adult users can be attributed to the use of smokeless tobacco. The risk of developing these leukoplakic lesions increases with increased exposure, and a number of studies now suggest that some snuff-induced leukoplakias can undergo transforma- tion to dysplasia and further to carcinoma. The evidence concerning the adverse health effects of smokeless tobacco use on other oral soft and hard tissues is only suggestive at this time. The magnitude of blood nicotine levels resulting from smokeless tobacco use has been shown to be similar to that from cigarette smok- ing. Therefore, the nicotinerelated health consequences of smoking would also be expected to result from smokeless tobacco use. Given the nicotine content of smokeless tobacco, the user's ability to sustain elevated blood levels of nicotine, and the wellestablished data implicat- ing nicotine as an addictive substance, it is reasonable to expect that smokeless tobacco is capable of producing nicotine addiction in users. There is also some suggestive evidence that nicotine may play a con- tributory or supportive role in the development of coronary artery and peripheral vascular disease, hypertension, peptic ulcer disease, and fetal mortality and morbidity. The conclusions in this report on the relationship between smokeless tobacco use and cancer, noncancerous and precancerous oral conditions, and addiction and dependence are substantially in agreement with those published at a recent National Institutes of Health (NIH) Consen- sus Development Conference on the Health Implications of Smokeless TRhacco use (12). CONCLUSIONS Prevalence and Trends of Smokeless Tobacco Use in the United States 1. Recent national data indicate that over 12 million persons used some form of smokeless tobacco (chewing tobacco and snuff) in 1985 and that approximately 6 million used smokeless tobacco weekly or more often. Use is increasing, particularly among young males. 2. The highest rates of use are seen among teenage and young adult males. A recent national survey indicates that 16 percent of males between 12 and 25 years of age have used some form of smokeless tobacco within the past year and that from onethird to onehalf of these used smokeless tobacco at least once a week. Use by females of all ages is consistently less than that of males; about 2 percent have used smokeless tobacco in the last year. 3. State and local studies corroborate the national survey findings. The prevalence of smokeless tobacco use by youth and young adults varies widely by region, but use is not limited to a single re gion. In several parts of the country, as many as 25 to 35 percent of adolescent males have indicated cnrrent use of smokelees tobacco. xxii Carcinogenesis Associated With Smokeless Tobacco Use 1. The scientific evidence is strong that the use of smokeless tobacco can cause cancer in humans. The association between smokeless tobacco use and cancer is strongest for cancers of the oral cavity. 2. Oral cancer has been shown to occur several times more fre- quently among snuff dippers than among nontobacco users, and the excess risk of cancers of the cheek and gum may reach nearly fiftyfold among long-term snuff users. 3. Some investigations suggest that the use of chewing tobacco also may increase the risk of oral cancer. 4. Evidence for an association between smokeless tobacco use and cancers outside of the oral cavity in humans is sparse. Some investigations suggest that smokeless tobacco users may face in- creased risks of tumors of the upper aerodigestive tract, but results are currently inconclusive. 5. Experimental investigations have revealed potent carcinogens in snuff and chewing tobacco. These include nitrosamines, poly- cyclic aromatic hydrocarbons, and radiation-emitting polonium The tobacco-specific nitrosamines N-nitrosonornicotine and 4-(methylnitrosamino)-l-(3-pyridyl)-1-butanone have been detected in smokeless tobacco at levels 100 times higher than the regulated levels of other nitrosamines found in bacon, beer, and other foods. Animals exposed to these tobacco-specific nitro samines, at levels approximating those thought to be accumu- lated during a human lifetime by daily smokeless tobacco users, have developed an excess of a variety of tumors. The nitro samines can be metabolized by target tissues to compounds that can modify cellular genetic material. 6. Bioassays exposing animals to smokeless tobacco, however, have generally shown little or no increased tumor production, although some bioassays suggest that snuff may cause oral tumors when testsd in animals that are infected with herpes simplex virus. Noncancerous and Precancerous Oral Health Effects Associated With Smokeless Tobacco Use 1. Smokeless tobacco use is responsible for the development of a por- tion of oral leukoplakias in both teenage and adult users. The degree to which the use of smokeless tobacco affects the oral hard and soft tissues is variable depending on the site of action, type of smokeless tobacco product used, frequency and duration of use, predisposing factors, cofactors (such as smoking or concomitant gingival disease), and other factors not yet determined. . . . xxm 2. Dose response effects have been noted by a number of investiga- tors. Longer use of smokeless tobacco results in a higher preva- lence of leukoplakic lesions. Oral leukoplakias are commonly found at the site of tobacco placement. 3. Some snuff-induced oral leukoplakic lesions have been noted upon continued smokeless tobacco use to undergo transforma- tion to a dysplastic state. A portion of these dysplastic lesions can further develop into carcinomas of either a verrucous or squamous cell variety. 4. Recent studies of the effects of smokeless tobacco use on gingival and periodontal tissues have resulted in equivocal findings. While gingival recession is a common outcome from use, gingivitis may or may not occur. Because longitudinal data are not available, the role of smokeless tobacco in the development and progression of gingivitis or periodontitis has not been confirmed. 5. The evidence concerning the effects of smokeless tobacco use on the salivary glands is inconclusive. 6. Negative health effects on the teeth from smokeless tobacco use are suspected but unconfirmed. Present evidence, albeit sparse, suggests that the combination of smokeless tobacco use in individ- uals with existing gingivitis may increase the prevalence of dental caries compared with nonusers without concomitant gingivitis. Reports of tooth abrasion or staining have not been substantiated through controlled studies; only case reports are available. Nicotine Exposure: Pharmacokinetics, Addiction, and Other Physiologic Effects 1. The use of smokeless tobacco products can lead to nicotine depen- dence or addiction. 2. An examination of the pharmacokinetics of nicotine (i.e., nicotine absorption, distribution, and elimination) resulting from smoking and smokeless tobacco use indicates that the magnitude of nico- tine exposure is similar for both. 3. Despite the complexities of tobacco smoke self-administration, systematic analysis has confirmed that the resulting addiction is similar to that produced and maintained by other addictive drugs in both humans and animals. Animals can learn to discriminate nicotine from other substances because of its effects on the cen- tral nervous system. These effects are related to the dose and rate of administration, as is also the case with other drugs of abuse. 4. It has been shown that nicotine functions as a reinforcer under a variety of conditions. It has been confirmed that nicotine can xxiv function in all of the capacities that characterize a drug with a liability to widespread abuse. Additionally, as is the case with most other drugs of abuse, nicotine produces effects in the user that are considered desirable to the user. These effects are caused by the nicotine and not simply by the vehicle of delivery (tobacco or tobacco smoke). 5. Nicotine is similar in all critical measures to prototypic drugs of abuse such as morphine and cocaine. The methods and criteria used to establish these similarities are identical to those used for other drugs suspected of having the potential to produce abuse and physiologic dependence. Specifically, nicotine is psychoactive, producing transient dose-related changes in mood and feeling. It is a euphoriant that produces doserelated increases in scores on standard measures of euphoria. It is a reinforcer (or reward) in both human and animal intravenous self-administration para- digms, functioning as do other drugs of abuse. Additionally, nice tine through smoking produces the same effects, and it causes neuroadaptation leading to tolerance and physiologic depen- dence. Taken together, these results confirm the hypothesis that the role of nicotine in the compulsive use of tobacco is the same as the role of morphine in the compulsive use of opium derivatives or of cocaine in the compulsive use of coca derivatives. 6. The evidence that smokeless tobacco is addicting includes the pharmacologic role of nicotine dose in regulating tobacco intake; the commonalities between nicotine and other prototypic dependenceproducing substances; the abuse liability and depen- dence potential of nicotine; and the direct, albeit limited at present, evidence that orally delivered nicotine retains the characteristics of an addictive drug. 7. Several other characteristics of tobacco products in general, in- cluding smokeless tobacco, may function to enhance further the number of persons who are afflicted by nicotine dependence: nicotinedelivering products are widely available and relatively inexpensive; and the self-administration of such products is legal, relatively well tolerated by society, and produces minimal d&-up- tion to cognitive and behavioral performance. Nicotine produces a variety of individual-specific therapeutic actions such as mood and performance enhancement; and the brief effects of nicotine ensure that conditioning occurs, because the behavior is associ- ated with numerous concomitant environmental stimuli. 8. All commonly marketed and consumed smokeless tobacco prod- ucts contain substantial quantities of nicotine. The nicotine is delivered to the central nervous system in addicting quantities when used in the fashion that each form is commonly used (or as recommended in smokeless tobacco marketing campaigns). XXV 9. Since the exposure to nicotine from smokeless tobacco is similar in magnitude to nicotine exposure from cigarette smoking, the health coIlSequences of smoking that are caused by nicotine also would be expected to be hazards of smokeless tobacco use. Areas of particular concern in which nicotine may play a contributory or supportive role in the pathogenesis of disease include coronary artery and peripheral vascular disease, hypertension, peptic ulcer disease, and fetal mortality and morbidity. REFERENCES (1) Christen, A.G., Swanson, B.Z., Glover, E.D., and Henderson, A.H. Smokeless tobacco: The folkIore and social history of snuffing, sneez- ing, dipping, and chewing. J. Am. Dent. Assoc. 105: 821829, 1982. (2) Gottsegen, J.J. Tobacco. A Study of Its Consumption in the United States. New York, Pitman, 1940, p. 3. (3) Voges, E. The pleasures of tobacco-How it ah began and the whole story. Tobacco J. Int. 1: 80-82, 1984. (4) Axton, W.F. Tobacco and Kentucky. Lexington, University Press of Kentucky, 1975, pp. 8, 25, 58-59. (5) Redmond, D.E. Tobacco and cancer: The first clinical report, 1761. N. Engl. J. Med. 282: 18-23, 1970. (6) Abbe, R. Cancer of the mouth. New York Med. J. 102: l-2,1915. (7) Ahblom, H.E. Predisposing factors for epitheliomas of the oral cavity, larynx, pharynx, and esophagus. Acta Radial. 18: 163-185, 1937 (in Swedish). (8) FriedeII, H.L.. and Rosenthal, L.M. The etiologic role of chewing tobacco in cancer of the mouth. JAMA 116: 2130-2135, 1941. (9) Moore, G.E., Bissinger, L.L., and ProehI, E.C. Tobacco and intraoral cancer. Surg. Forum 3: 685-688, 1952. (10) U.S. Public Health Service. Smoking and Health. A Report of the Surgeon General. Department of Health, Education, and Welfare, Public Health Service, Office of the Assistant Secretary for Health, Of- fice on Smoking and Health (DHEW Publication No. PHS 79-50066). Washington, D.C.. U.S. Government Printing Office, 1979, pp. 13-38 to 13-41. (11) International Agency for Research on Cancer. Tobacco habits other than smoking: Betel-quid and areca-nut chewing and some related nitrosamines. IARC Monogr. 37: 291,1985. (12) National Institutes of Health. Consensus Development Conference Statement on the Health Implications of Smokeless Tobacco Use, January 13-15, 1986. xxvi Chapter 1. PREVALENCE AND TRENDS OF SMOKELESS TOBACCO USE IN THE UNED STATES CONTENTS Introduction ..................................... ...5 Product Characteristics. .............................. .5 l'kendsinProductionandSaIes ......................... .5 Categories of Products ............................ .7 Tbmporallkends ............................... ...7 PendsinSeIf-ReportedUse: SurveyData. ............... .7 National Survey Data ............................. .7 StateandLocaISurveyData ...................... .15 Conclusions........... . ..24 ResearchNeeds.....................................25 References.........................................26 3 INTRODUCTION This chapter defines the various forms of smokeless tobacco that are used in the United States and e xamines the data that pertain to trends in prevalence and patterns of use. ?kends in smokeless tobacco produc- tion and sales and self-reported use are considered. Methodological con- siderations are discussed and research needs are identified. Tobacco was used by pre-Columbian American Indians in smokeless forms as well as smoked (1). Cultivated by American colonists, tobacco became a major commodity in trade with Europe. Until the end of the 19th century, the use of smokeless tobacco products was widespread in the United States. Its use declined rapidly in this century with the advent of antispitting laws, loss of social acceptability, and increased popularity of cigarette smoking (1,2). Use was primarily confined to rutd and agricultural areas and to occupational settings where smok- ing was not aIlowed, such as mining and some industries (3,4). In the Southeastern United States, especially in rural areas, oral use of dry snuff remained pop&r among women (5,6). PRODUCT CHARACTERISTICS T&lay, smokeless tobacco is produced in two general forms: chewing tobacco and snuff (7-10). Chewing tobacco is chewed or held in the cheek or lower lip. Three primary types of chewing tobacco are marketed: looseleaf, plug, and twist. Snuff has a much finer consistency than chewing tobacco and is held in place in the mouth without chewing. It is marketed in both dry and moist forms. Although smokeless tobacco is not subject to combustion and is usually used orally in the United States, products differ with regard to several factors, including type of tobacco plant used, parts of the tobacco plant used, method of curing, moisture content, and additives. For example, looseleaf chewing tobacco is made from aircured, cigar-type leaves from tobacco that is grown in Pennsylvania and Wisconsin In contrast, dry snuff is made primarily from firecured dark tobacco that is grown in Kentucky and `Ibnnessee. Plug tobacco and snuff come in dry and moist forms. Many smokeless tobacco products are sweetened with sugar or molasses. Many are flavored; licorice is a common additive for chewing tobacco, while mint and wintergreen often are used to flavor snuff. able 1 describes the types of smokeless tobacco and how they are used and packaged (7-101. TRENDS IN PRODUCTION AND SALES United States Department of Agriculture (USDA) records on the annual production and sales of smokeless tobacco serve as indicators of the population's consumption Changes in consumption can be inferred from changes in production and sales. Because sales figures closely resemble those for production, only production will be reported. 5 0.J TABLE l.-Characteristics of Smokeless Tobacco Products Product Description How used Packaging* Plug `I\vist CHEWING TOBACCO Made from airwred. cigar leal tobaccos of I'ennsylvan~a and Wisconsin. Consists 111 stripped and procrssed Lobacco leaves. The leaves are sttvnmtul. rut. or granulated and are loosely packed to form small strips of shredded tobacco. Most brands are sweetened and flavortxl with licorice. Made from enriched tobacco leaves (I~urlry and bright tobacco and cigar tobacco) or fragments wrapped in fine tobacco and pressed into bricks. May be firm (less than 15 percent moisture) or m&l (15 percent or greater moisture). Most plug tohacro is swwlened and flavored with licorice. Handmade of dark, air-cured leaf tobacco treated with a tarlike tobawo leaf extract and twisted into strands that are dried. Majority is sold without flavoring and sW~La"ars. A piece of tobacco. 314 Lo 1 inch in diameter, is tucked between the gum and jaw, usually Lo the back of the mouth. Pouch. typically 3 ounces. A few brands market a 1.5-ounce pouch. Chewed or held in the cheek or lower lip. May be held in the mouth for several hours. A compressed brick or flat block wrapped inside natural tobacco leaves. Packaged in clear plastic. Packages range from 7 Lo 13 ounces. Also sold by the piece. Similar to plug. A pliable but dry rope. Sold by the piece, packaged in plastic bags. No standard weight. Sold in small (approximately l-2 ounces) and larger sizes based on the number of leaves in the twist. SNUFF Moist Dry Made from air-cured and fire-cured tobacco. Consists of tobacco sLems and leaves that are processcld into fine par- ticks or strips. Some products are flavored. Has a moisture content of up to 50 percent. Most dry snuff is made from fire-curtd tobaccos of Ken- tucky and Tennessee. After initial curing. the tobacco is fermented further and processed into a dry powdered form. Products vary in strength and flavoring. tienerally has a moisture content of less than 10 percent. A small amount ("pinch") is placed between the lip or cheek and gum and is typically held for 30 minutes or longer per pinch. Same as moist snuff. May also be sniffed. Cans and plastic containers. typically 1.2 ounces. Metal cans or glass containers, vary from 1.15 to 7 ounces per container. * Product weight Iincludes moisture). Categories of Products The USDA reports production and sales by product category (i.e., chewing tobacco and snuff). The definitions of categories changed in 198 1. Prior to 198 1, total figures for chewing tobacco were derived by summing data for the subcategories of plug, twist, looseleaf, and fine cut; snuff was a separate category. However, finecut tobacco is used in moist snuff. `Ih reflect this fact, after 1981 USDA shifted fine-cut from the category of chewing tobacco to moist snuff. Tb observe and clarify temporal trends for the purposes of this review, the data presented in figure 1 reflect a uniform category system across years. In these records, finecut tobacco is counted consistently as snuff (11-17). Temporal Trends Figure 1 depicts temporal trends in the quantities of smokeless tobacco that were manufactured in the United States from 1961 to 1985. Be tween 1944 and 1968, total smokeless tobacco production declined 38.4 percent from 150.2 to 92.5 miI.Iion pounds. Subsequent increases in pro duction reached 135.6 rniIIion pounds in 1985. Between 1970 and 1985, total snuff production increased 56 percent from 31.3 to 48.7 million pounds. This increase was due to changes in the production of moist snuff; the manufacture of dry snuff dechned (3). The difference in trends in the production of moist and dry snuff is shown in figure 1 for the years 1981 through 1985. Separate production data are not available for the two types of snuff prior to 1981. Between 1970 and 1981, however, the production of finecut tobacco, used in the manufacture of some moist snuff, increased threefold from 4.8 to 15.2 miIIion pounds. Between 1970 and 1985, the production of chewing tobacco increased 36 percent from 63.9 to 86.9 miIIion pounds. This increase was due to the production of looseleaf tobacco, which increased 87.3 percent from 39.5 to 74.0 mihion pounds. The production of plug and twist tobacco declined during this period. National Survey Data National data from 1964 to 1985 are available from eight different na- tional probability surveys and a national survey of college students. The majority of the data pertain to persons over the age of 17. The prin- cipal characteristics of these surveys are shown in table 2. Office on Smoking and He&h Surveys Early data on the use of chewing tobacco and snuff are available from the 1964.1966.1970, and 1975 Adult Use of Tobacco Surveys that were FIGURE l.-Manufacturing lhdsz Quantities of Smokeless `Ibbacco Mamdactured in the United States From 1961 to 1985 Expressed in Million Pounds o ? ?? ?? ? ? ? TABLE 2.-National Prevalence of Smokeless Tobacco Use: Data Sources Number of survey * Date Respondents Respondents/ Households Products Questions Office on Smoking and Health Office on Smoking and Health Office on Smoking and Health Oifice on Smoking and Health National Health Interview Survey Supplement (National Center for Health Statistics) Simmons Study of Media Markets, Simmons Market Research Bureau, Inc. Simmons National College Study, Simmons Market Research Bureau, Inc Personal Interview Personal Interview Telephone `IbIephone Personal Interview Including Proxy Questionnaire Questionnaire Current Population Survey Personal Supplement-Census Bureau Interview for Office on Smoking I"chJdi"g and Health Proxy NIDA Household P~sO"Ed Survey Interview 1964 Adults L 21 5,794 1966 Adults L 21 Adults 2 21 Adults > 21 Persons 2 17 5,770 1970 5,200 1975 1970 12.000 77,ooo/ 37.ooo 1980 1981 1982 1983 1984 1985 1983 1985 Adults 2 18 15,ooo- 19,ooo Cdegs Students 2 18 2,011- 2,373 1985 Persons 1 16 12o.o00/ Snuff and Chewing 58,000 Tobacco Separately 1985 Persons > 12 8,ooO Snuff and Chewing Tbbacco Separately Snuff and Chewing Tobacco Separately Snuff and Chewing Tbbacco Separately Snuff and Chewing -Ibbacco separately Snuff and Chewing lbbacco Separately Snuff only Snuff only Snuff and "On the average. in the past 12 months, Chewing how often have you used chewing tobacco `Ibbacco Combined or snuff or other smokeless tobacco?" "Have you ever used-at aII regularly?" "Do you use-now?" "Have you ever used-at aII regularly?" "Do you use-now?" "Have you ever used-at all regularly?" "Do you use-now?" "Have you ever used-at alI regularly?" "Do you use-now?" Dol?S presently use any other form of smokeless tobacco, such as snuff or chewing tobacco? 1980 to 1983 "Do you use it yourself- snuff (smokeless tobacco)?" 1984 to 1985 "Do you yourself use any of the following tobacco products?" Snuff (ST) listed as an option. "Please mark which of the items listed below you yourself use." Snuff (smokeless tobacco) listed as a" option. Does presently use any other form of tobacco. such as snuff or chewing tobacco? What other forms of tobacco does presently use? TABLE 3.-Use of Smokeless Tobacco in the United States by Individuals Over 21 Years of Age* Percentage of Users Males Females Use Catesrw 1964 l!x6 1970 1975 1964 1966 1970 1915 Now Use Snuff 2.0 3.1 2.9 2.5 2.0 2.1 1.4 1.3 Used to Use Snuff 3.6 3.9 4.2 4.0 0.9 1.0 1.1 1.1 Have Ever Used Snuff t 5.7 7.2 7.1 6.4 2.9 3.1 2.6 2.4 Now Use Chewing Tobacco 5.1 7.1 5.6 4.9 0.5 0.4 0.6 0.6 Used to Use Chewing mbacco 12.0 13.2 19.1 16.1 1.0 1.1 1.8 1.2 Have Ever Used Chewing Tobaccot 17.2 20.5 24.7 21.0 1.5 1.5 2.4 1.8 * "Use" not further defined with respect to frequency. t Includes those who used to use. but did not state if they used it currently. Source: National Clearinghouse on Smoking and Health conducted by the National Clearinghouse for Smoking and Health, cur- rently the Office on Smoking and Health (OSH) (18-20). National prob ability samples of 5,700 to 12.000 individuals over the age of 21 from randomly selected households were interviewed by telephone regarding the use of tobacco products. Between 1964 and 1975, the prevalence of smokeless tobacco use remained fairly stable. Results are summ&zed in table 3. Three patterns in these data may be noted: o Less than 5 percent of the population reported using smokeless tobacco. o Nationally, use was higher among males than females. o Among males, the prevalence of use of chewing tobacco was higher than that for snuff. National Health Interview Survey In 1970, the National Center for Health Statistics included a question on current use of snuff and chewing tobacco in its National Health Interview Survey (NHIS) (21). One respondent per household provided information on alI household members age 17 and older. Data were col- lected on approximately 77,000 persons in 37,000 households. Esti- mates indicate that 1.4 percent of males used snuff and 3.8 percent used chewing tobacco (table 4). Simmons Market Research Bureau, Inc. National probability data that were coIIected annuaIIy from 1980 through 1985 for the Simmons Study of Media and Markets provide estimates of the prevalence of snuff use among adults who were 18 years of age or older. Sample size ranged from 15,000 to 19,000. Data are ed in table 5 for the years 1980 to 1985. The prevalence 10 TABLE 4.-Prevalence of the Use of Snuff and Chewing `lbbncco Among Malee by Age, 1970 NHIS and 1985 CPS Surveys* 1970 HIS 1985 CPS Product Snuff Chewing Tobacco Age 17-19 20-29 30-39 40-49 50+ Total 17-19 20-29 30-39 40-49 50+ lbtal Percentage of usf!m 0.3 0.6 0.7 1.2 2.7 1.4 1.2 1.9 2.8 3.0 6.5 3.8 Age 16-19 20-29 30-39 40-49 50+ ?btal 16-19 20-29 30-39 40-49 50+ mal Percentage of uaem 2.9 2.7 1.8 1.5 1.4 1.9 3.0 4.2 3.7 3.3 4.2 3.9 o "Use" *?? further defined with respect to frequency. Sources: National Center for Health Statistics. National Health Interview Survey, 1970 (unpublished). Office on SmoLing and Health, Current Population Survey. 1985 (unpubtiahedl. TABLE B.-National Prevalence of Current Use of Snuff by Gender, Age, and Race for 19&o Through 1985* Percentage of Ufsera Sample 1980 1961 1982 1963 1984 1985 Total 1.6 2.2 2.6 2.3 1.9 1.9 Gender Males Females 2.4 3.7 4.2 3.8 3.0 3.2 0.8 0.8 1.1 0.9 1.0 0.7 Ali!= 18-24 25-34 35-44 45-54 55-64 2 65 1.4 2.6 4.3 3.5 3.2 2.8 2.5 2.8 3.1 3.0 2.0 2.1 1.07 1.3 1.6 1.8 1.5 1.0 1.37 1.3 1.47 1.07 1.17 1.5 1.2t 1.7 1.7 2.3 1.1t 1.3 1.67 2.8 2.6 1.4 2.5 2.4 Race Black White Other 2.3t 1.6t 3.0 2.9 2.9 2.4 1.5 2.2 2.6 2.3 1.9 1.9 1.97 1.47 1.17 NA 0.4t 1.2 o Adults defined as individuals over 18 years of age. Use not further defined with respect to frequency. t Number of cases too small for reliable estimates. Source: Simmons Market Reaemch Bureau. Inc.. Study of ????? and Markets. 1980.1985. 11 TABLE 6.-Prevalence of Snuff Use Among College Students 18 Years of Age or Older by Gender and Year* Percentage of Users Sample 1983 1986 Total Gender Males Females Race Black White Other 2.7 3.5 5.4 6.7 0.17 0.2f 1.5t 1.4-f 5.1 3.6 4.97 4.37 o Current use; frequency of "se not specified. t Projection relatively unstable because of small sample. Source: Simmons Market Research Bureau, Inc.. Simmons National College Study, 1983 and 1985 rate for "current use" of snuff was 2.4 percent for males in 1980 and 0.8 percent for females. Rates for males peaked at 4.2 percent in 1982 and were 3.2 percent in 1985. Since 1982, the highest rates of use have con- sistently been observed in the age group 18 to 24 years old. Compara- tively higher rates of use were also observed in the age groups 25 to 34 years old and over age 65 (22. The Simmons National College Study reports data from a probability sample of full-time students 18 years or older who were attending baccalaureategranting colleges and universities in the coterminous United States. In 1983, 2,011 students were sampled, and 2,373 students were sampled in 1985. Five to 7 percent of males indicated use of snuff compared to 0.2 percent of females (table 6). The prevalence rate among male students exceeded that of the general adult male popula- tion (tables 5 and 6). In 1985, prevalence among college males was twice that of other adult males, while the rate for college women was less than onethird that among the general adult female population. The com- bined prevalence for male and female college students (3.5 percent) was very similar to that for 18- to 24yearolds in the general population (2.8 percent) (tables 5 and 6) (23). Current Population Survey In the fall of 1985, the Census Bureau collected health information on approximately 120,000 persons in 58,000 households in its Current Population Survey (CPS) (24). OSH sponsored a supplement to this survey, which included a question on current use of snuff and chewing tobacco. One respondent per household provided information on all members age 16 and older. Provisional estimates of smokeless tobacco use indicate that 1.9 percent of males used snuff and 3.9 percent used chewing tobacco (table 4). 12 TABLEI 7.-National Prevalence of Smokeless `Ibbaccu Use by Adult Status aud Sex, NIDA Sample, 198!5* Percentage. of Users Males Females Use Category 12OYears 2 21 Years I 20 Years r21Years Used in Past Year 16 11 2 2 Used Formerly 4 7 2 2 Never Used 79 82 96 96 o Preliminary estimates not adJusted for oversampling of blacks and Hispamcs. Source: National Institute on Drug Abuse, 1985 Natmnal Household Survey on Drug Abuse Preliminary results presented at the NIH Consensus Development Conference on the Health Implications of Smokeless lbbacco Use. January 1986. TABLE 8.-Recency of Smokeless Tobacco Use by Sex and Age Group* Percentage of Users by Age Groups 12-17 l&25 26-3s 40-t Use Categom Males Females Males Females Males Females Males Females Usedin Past Year 16 1 16 1 10 1 8 3 Used Formerly 4 2 7 1 5 1 8 2 Never Used 80 97 77 98 85 98 84 95 * P&mmary estimates not adjusted for oversampling of blacks and Hispanics Source: National Institute on Dtug Abuse, 1985 National Household Survey on Drug Abuse. Preliminary results t; resented at the NIH Consensus Development Conference on the Health Implications of Smokeless Tobacco Use. anuary 1986. National Institute on Drug Abuse Housebold Survey The recently completed 1985 National Household Survey on Drug Use provides the national probability data on current use and correlates of use of smokeless tobacco by youth It is the eighth in a series of na- tional probability surveys conducted among household residents in the coterminous United States by the National Institute on Drug Abuse (NIDA). Data are collected on the use and adverse consequences that are associated with 11 drugs or drug classes. The 1985 survey over- sampled for blacks and Hispanics and younger age groups. The total sample consists of approximately 8,000 facet-o-face interviews. The data presented here are based on a prehminary analysis of 4,564 inter- views. provisional estimates are presented in tables 7 through 9. Sixteen percent of males under the age of 21 reported using chewing tobacco or snuff within the last year, in contrast to 11 percent of older males (table 7). The decline in older age groups is seen more clearly when narrower age categories are used (table 8). An estimate of the preva- lence of weekly use may be obtained by combining the use frequency 13 TABLE 9.--Frequency of Smokeless Tobacco Use in Past Year* Percentage of Users Past Year Use of Smokeless Tobacco Age Groups for Males 12-17 18% 2&39 40+ Males and Females Age 12 and Above Most Days/Week 3 7 5 4 2 1 or 2 Days/Week 2 1 1 1 1 1 or More Days/Week 5 8 6 5 3 3-51 Days/Year 5 5 3 3 2 1-2 Days/Year 6 3 2 1 2 Not in Past Year 4 7 5 8 3 Have `Itied 20 23 15 16 10 Never 80 77 85 84 90 o Prelmnary estunates not adJusted for oversampling of blacks and Hispanics. Source National Institute on Drug Abuse. 1985 National Household Survey on Drug Abuse. Preliminary results presented ar. the N IH Consensus Development Conference on the Health Implications of Smokeless Tobacco Use. January 1986. categories of "most days a week" and "1 or 2 days a week" (table 9). Use at least once a week peaks in the 1% to 25year-old age groups at 8 per- cent. As in previous surveys, the use among females was consistently much lower than among males. Responses suggest slightly higher rates of use among women 40 years of age and older than among younger women (table 8) (25). Discussion of National Survey Data Despite varying methodologies among the national surveys (table 2), sufficient commonalities permit mean@ful comparisons. The 1970 and 1975 OSH surveys and the 1980 to 1985 Simmons Study of Media and Markets indicate that the use of snuff by adult males remained con- stant within a range of 3 to 4 percent. Use by adult females also re- mained constant at about 1 percent. During this same l&year period, the population over the age of 18 increased 32 percent from 133.5 million to 175.8 million (26). The production of all forms of smokeless tobacco increased 42 percent from 95.2 to 135.6 million pounds, and the production of finecut/moist snuff tripled. This may indicate the emergence of a new population of users. The 1970 NHIS and the 1985 CPS both relied on the use of proxy re spondents. Estimates of smokeless tobacco use are likely to be lower than the actual population prevalence because respondents may not always be aware of smokeless tobacco use by other members of the household. In fact, in 1970, the NHIS estimated that 1.4 percent of males used snuff and 3.8 percent used chewing tobacco. In the same year, the OSH Adult Survey, which did not use proxy respondents, pro vided corresponding estimates of 3 and 6 percent. Similarly, the CPS estimates that 1.9 percent of males used snuff in 1985, while the Sim- mons Study of Media and Markets estimates 3.2 percent. 14 However, comparisons between the 1970 NHIS and the 1985 CPS for the purpose of e xamining trends are appropriate. They suggest little change in the overall rate of adult male use of smokeless tobacco but indicate a marked change in the age distribution of users (table 4). In 1970, the use of smokeless tobacco was most common among older men; in 1985, the prevalence in the younger age groups had greatly increased. Both the Simmons Study of Media and Markets and the NIDA survey show the highest rates of use among young adults ages 18 to 24. The Simmons National College Study indicates that male college students are as likely to use snuff as are other 18- to 24-year-olds. The Simmons data also show a slight elevation in prevalence among persons over the age of 65, which reflects the age distribution of traditional users of smokeless tobacco. If the NIDA prevalence estimates are applied to current population figures (261, there are at present over 12 million persons in the United States ages 12 and older who have used some form of smokeless tobacco in the past year. Three million are under the age of 21, and 1.7 milhon of these are males 12 to 17 years old. An estimated 6 million persons use smokeless tobacco at least weekly. Of these, 0.5 million are males ages 12 to 17; 1.3 million are males ages 18 to 25; and approximately 780,000 are females. The 1980 to 1985 Simmons Study of Media and Markets estimated that 2 to 4 million persons over the age of 18 were users of snuff. Of these, 0.6 to 1.2 million were between the ages of 18 and 24. able 10 summar%es data on the prevalence of smokeless tobacco use by region from three national surveys conducted in 1985. Among these adult samples, use was highest in the South and lowest in the North- east, with the West and North Central/Midwest falling in between. These surveys provide self-report data only; no direct validation at- tempts were made. Because no strong social sanctions regarding smokeless tobacco use exist for adults, systematic misrepresentation by them is unlikely. However, under the conditions of a personal inter- view, as used in the NIDA study, adolescents would be more likely to underreport than over-report their use of smokeless tobacco. In addi- tion, the prehminary estimates from the NIDA survey have not been adjusted for oversampling of blacks and Hispanics. In this sample, blacks and Hispanics reported less smokeless tobacco use than whites, and their over-representation would result in underestimates of national prevalence. State and Local Survey Data State and local surveys provide much of the information after 1980 on the use of smokeless tobacco. Since most of these surveys were con- ducted in schools, often motivated by apparent increases in students' 15 TABLE lo.-Prevalence of Smokeless Tobacco Use by census Region, 1985 Prevalence Category Percentage Reporting Use Northeast North Central South West CPS Chewing Tobacco Snuff Simmons Snuff NIDA* (Snuff and/or chewing tobacco) Weekly Use or More Often Any Use in Past Year 1.6 3.7 7.0 3.9 1.2 2.3 3.1 1.6 1.5 1.3 2.9 1.3 1.0 2.0 5.0 4.0 4.0 6.0 8.0 9.0 * Preliminary estimates not adjusted for age and race Sources: Office on Smoking and Hex&b. Current Population Survey. 1985 iunpublished). Simmons Market Research Bureau. Inc., Study of Media and Markets. 1980-1985. National Institute on Drag Abuse, 1965 House- hold Survey on Drug Abuse. Preliminary results presented at the NIH Consensus Develqxnent Conference on the Health Implications of Smokeless lbbscco Use, January 1986. use of smokeless tobacco products, there may be a selection bias. However, the large and growing number of reports and the wide geographic coverage support the conclusion that smokeless tobacco use is not a localized phenomenon, Indeed, the consistency of such data sug- gests that smokeless tobacco has become a product that is used by large numbers of teenage and young adult males. Adult Use Several reports provide a tentative profile of local usage patterns of smokeless tobacco among adults. In 1979, tobacco use information was collated from 4,282 men between the ages of 21 and 84 in 10 geographic areas as part of the National Bladder Cancer Study, a population-based case control study (27). The overall prevalence for having "ever used snuff for 6 months or more" among the control subjects (randomly selected from the general population) was 5 percent; for chewing to bacco, the corresponding figure was 12 percent. A breakdown by age indicated much more use of smokeless products by older men than younger men (table 11). Glover and his colleagues conducted a random sample telephone survey of 280 persons in Pitt County, North Carolina (28). A user was defined as a person who answered "yes" to the question, "Do you dip or chew tobacco?" Forty percent of males and 9 percent of females answered positively. High rates of use are probably not a new phenome non since. there is a tradition of smokeless tobacco use among both sexes in this area, and tobacco is a major agricultural product,. 16 TABLE Il.--prevalence of Snuff and Chewing Tobacco Use by Adult Males in 10 Geographic Areaa Percentage Reporting Ever Used Sample n Snuff Chewing lbbacco All Men 4.282 5 12 Age 21-44 45-64 65-84 240 0 2 1,653 3 6 2,389 7 16 Area of Residence Atlanta Connecticut Detroit Iowa New Jersey New Mexico New Orleans San Francisco Seattle Utah 186 8 23 654 4 12 355 8 20 552 12 14 1.288 2 10 129 7 20 115 1 6 542 2 8 255 10 6 206 5 7 Race White Nonwhite 3,892 5 11 390 5 18 Source: National Bladder Cancer Study. Hartge. P.. Hoover. FL. and Kantor. A. Bladder cancer risk and pipes. cigars. and smokeless tobacco. Cancer. 55: 901-906. 1985. Research supported by the National Cancer Institute, the Food and Drug Administration. and the Environmental Protection Agency. Gritz, Ksir, and McCarthy surveyed a sample of 214 students at the University of Wyoming (29). In their sample, 27.1 percent of males and 4.1 percent of females reported "current use," with the criterion for "current use" unspecified. The vast majority of users (84 percent) used moist snuff. Glover and his colleagues reported a survey of 5,894 students in physical education classes at 72 colleges and universities from 8 States (Oregon, Arizona, Colorado, Oklahoma, Minnesota, Ohio, South Carc~ lina, and Connecticut) (30). `l%enty-two percent of the males who were surveyed reported using smokeless tobacco compared to 2 percent of the females. Combined rates of use for both sexes ranged from 15 per- cent in Oklahoma to 8 percent in Connecticut. The majority of the users reported using less than one can or pouch per week. Adolescent Use Studies of school-age youth conducted since 1980 are summarized in table 12 13145). Five different criteria for classifying use have been selected for data display: daily use, weekly use, monthly use, current use (no frequency specified), and ever used. Recent regional data on the use of smokeless tobacco have been col- lected by a number of National Cancer Institute grantees in the course 17 TABLE EL-Prevalence of Use of Smokelese lbbacco Among Youth by Gender and Grade: Regional and StateLevel Surveys Reported since 1!Bo* LOC.StiOll keference~ Grade&$ Malea Females Total I Daily Use Arkansas (31) Arkansas (32) Nebraska (33) Ohio (34) Chewing Tobacco Snuff Oregon (35) Oregon (36) wisconsin (37) lo-12 lo-12 7-12 4-12 7 9 10 7 a 9 10 7 8 9 10 11 12 lbtal 26.0 - 2.5 11.4 19.7 a.8 18.5 23.1 4.6 5.8 9.7 10.6 3.0 6.0 3.0 8.0 11.0 15.0 - - - 0.0 0.2 0.4 0.7 0.0 2.4 - - - - 0.0 0.0 0.0 0.0 0.0 0.0 - - 179 15.0 901 - 2,612 - 1.004 - 1,004 - 443 - 249 - 130 - 710 - 139 - 432 - 255 - - - - - - - - - - weekly use (Or more often) Nebraska (33) wisconsin (37) 7-12 7 8 9 10 11 12 lbtal 4.8 0.0 - 2.616 12.0 - - 18.0 - - - 15.0 - - - 24.0 - - - 25.0 - - 37.0 - - - - 1.0 - 25,000 Monthly Use (Or more often) Arizona (38) Midwestern states (39) Nebraska (33) 8-12 18.4 - - 1,080 lo-12 33.0 0.0 - 323 7-12 7.1 0.0 - 2.616 Current Use (Frequency not specified) Arkansas (31) lo-12 Arkansas (32) 10 11 12 T&al 31.8 2.2 - 179 - - 13.8 326 - - 20.6 330 - - 23.7 245 36.7 2.2 - 901 18 TABLEIl2.-Continued LOtXtiOll (reference) Grade(s) M&Ii Females Total n Current Use (Cont.) Colorado (40) lo-12 21.6 0.6 - 1,119 Colorado (4 1) 10-12 26.0 0.0 - 445 Louisiana (42)f 1976-1977 Chewing Tobacco 8-9 11.0 - - - lo-11 17.0 - - - 12-13 25.0 - - - 14-15 24.0 - - - 16-17 15.0 - - - Snuff 8-9 4.0 - - - 10-l 1 7.0 - - - 12-13 5.0 - - - 14-15 11.0 - - - 16-17 5.0 - - - Total - - - 2.880 1981-1982 Chewing Tobacco 8-9 24.0 - - - IO-11 32.0 - - - 12-13 39.0 - - - 14-15 43.0 - - - 16-17 15.0 - - - Total - - - 1.981 Snuff 8-9 21.0 - - - lo-11 26.0 - - - 12-13 32.0 - - - 14-15 30.0 - - - 16-17 14.0 - - - lbtal - - - 1,981 Pennsylvania (43) 7-12 30.0 0.0 - 538 Texas (44) 7-12 19.0 0.0 - 5,392 Wyoming (29) 7-9 24.5 1.2 - 2,408 Ever Used Arkansas (45) K Ohio (34) Chewing Tobacco 4-12 lbtal Snuff 4-12 Total Oregon (35) 7 9 10 Wisconsin(37) 7 8 9 10 11 12 Total 58.0 - 64.0 - 63.4 72.7 76.7 32.0 45.0 47.0 50.0 47.0 48.0 - 12.0 - 24.0 - 19.9 16.4 23.8 - - 21.4 112 - - - 1,007 - - - 1,007 - 445 - 249 - 133 - - - - - - 11.0 25,000 o Unless otherwise indicated, figures represent the usa been made for studies that provide for more than one c E... of chewing tobacco andlor snuff. Multiple entries have sslfrcatmn cntenon. t Age listed rather than grade. 19 of their ongoing research on tobacco use by youth (4s). Through col- laboration, these investigators have achieved more standardization in data collection than in previous studies, which makes comparisons among the different locales more meaningful. Although there were some differences in methodology, all of the studies addressed one or both of the following research questions: 1. What percentages of males and females have ever used smokeless t&lCCO? 2. What percentages of males and females have used smokeless tobacco in the last 7 days? Adolescent males may be subject to pressures that simultaneously discourage and encourage smokeless tobacco use. Underreporting of use may result from the presence of teachers and the setting in which the survey is administered. Overreporting may result from peer pressure to be seen as a smokeless tobacco user. Accurate reporting may be facilitated by collecting breath or saliva samples when surveys are completed. Respondents who believe that their se&reports can be objectively verified via biochemical testing tend to provide more accu- rate responses (47-49). Biochemical validation was used in 14 of the 17 subsamples reported in table 13. Most studies do not distinguish between snuff and chewing tobacco. In reports where the two have been separated, both substances were found to be in use (344243). Rates of smokeless tobacco use were consistently higher among males than females. This difference is especially marked when more precise classifications for regular use are employed. While substantial numbers of adolescent females report having tried smokeless tobacco at least once, very few use it on a regular basis (3335,37,39,&J. The use of smokeless tobacco by youth was generally higher in rural than urban areas, in small communities, and in areas where there is a tradition of smokeless tobacco use (Z&$37,46). However, high rates of use have also been reported in large metropolitan areas as well (37,40,46). able 14 smmm&es data on smokeless tobacco use by ethnic groups collected by investigators using standardized questions (46). lb date, lit- tle information has been available on smokeless tobacco use by non- whites, and some early research suggested that minority youth were not taking up the practice (42). In these studies, however, Hispanic youth showed rates of smokeless tobacco use comparable to whites, and Native American rates were consistently higher. In most locales, use was less common among Asians and blacks. Nationally, black college stu- dents are less likely to use snuff than are white college students Itable 6). Prevalence estimates for smokeless tobacco use by black adults, however, have equaled or exceeded those of whites (tables 5 and 11). The likelihood of using smokeless tobacco appears to increase with age as well as over time (32-35,37,42,46). Only one study has collected 20 TABLE 13.-Prevalence of Use of Smokeless lbbacca Among Youth by Gender and Grade: Local Surveys Using stana Questions Sample Males Females Grade Percentage n Percentage Il Used in Last 7 Days California Suburban/Rural Minnesota Suburban/Urban Montana Urban New York Urban New York New York City New York Suburban Oregon Suburban/Rural Oregon Suburban/Urban Southeastern unitf!d states 10 SMSA's Vermont Rural Vermont Urban Washington Rural Washington RLUal 4.7 (469) 0.7 (407) 14.8 (574) 1.4 (557) 9.2 (487) 1.6 (499) 9 18.1 (2.015) 2.4 (2.146) 9.4 (477) 2.0 (4031 11.9 (429) 1.5 (392) 13.9 (446) 3.2 (402) 3.9 (306) 2.9 1272) 10.7 (252) 0.3 it: ww (275) (243) 6 1.1 (1,488) 0.9 U.494) 7 3.0 (2,016) 0.0 (1,811) 6 ii 9 10 11 E 13:6 17.3 22.2 22.7 (602) (627) (6631 (572) (514) (440) 0.9 0.8 it: 2:3 0.5 (542) (618) 608) (567) (471) (431) 6 7 ; 1.9 (571) 0.4 625) 4.6 (570) 1.4 (575) 6.8 (514) 0.8 (533) 14.8 (588) 1.2 (575) 6 9.8 (305) 1.3 ww 7 12.1 (346) 0.6 (325) 8 10.4 v79) 1.6 (313) 9.3 mw 0.3 (317) 14.9 (328) 1.0 (289) 4 2.8 (216) 0.0 w9) 5 4.8 (207) 1.0 cw 6 5.4 (204) 0.0 (193) 4 2: 6 8.8 7 13.1 8 14.8 (45) 0.0 (47) (141) 1.3 056) W8) 2.1 (964) (521) 4.1 (514) (316) 5.2 (325) 10 23.7 (215) 0.4 (233) 21 TABLE 13.-Continued Males Females Sample Grade Percentage P Percentage n Ever Used California Suburban/Rural California pG/;pples California Los Angeles SMART California Los Angeles TVSP Minnesota Suburban/Urban Montana Urban New York Urban New York New York City New York Suburban Oregon Suburban/Rural Oregon Suburban/Urban Southeastern united states 10 SMSA's Vermont Rural Vermont Urban Wzu$inl@n Waterloo. Canada Suburban/Rural 32.6 56.2 56.7 24.9 7.8 19.6 20.0 6.7 I:;:; (504) (310) 25.3 (479) 31.9 1429) ii:: 32.0 (1,240) 6.9 (480) (418) (1,474) 62.1 (2,001) 22.9 (2,133) 41.0 56.9 68.2 17.5 19.3 24.6 23.1 (307) 3.4 33.5 (272) 5.1 47.8 (255) 7.0 i275; (24.3) 6.7 (1,488) 3.0 u,494 25.3 (2.016) 4.1 11,811) 48.3 (607) 16.2 57.9 639) 19.8 64.5 (677) 23.8 70.4 (577) 26.7 74.7 (5221 31.1 77.5 (445) 34.2 (551) I:?;; (576) (4851 (436) 32.4 (568) 44.9 (568) 54.1 (51.2) 61.3 1589) E 17:2 24.7 (528) f%J (575) 47.6 49.0 51.4 11.4 ew 13.5 (325) 15.6 (314) 38.8 1289) 8.2 (317) 54.8 (332) 7.2 (290) 17.4 (213) 26.2 (207) 39.8 (206) %!I 3.1 l%1 (193) 15.6 27.0 49.0 52.0 58.9 145) (141) 1E1 (316) 0.0 (47) 7.7 (156) 13.0 (964) 16.0 (514) 20.1 (325) 73.5 (215) 26.0 (281) 30.9 5.5 WW (444) 22 TABLE 14.-Mean Frequency of Smokeless Tihacco Use During Last 7 Days by Ethnicity of Male Respondents Sample California Suburban/Rural Grades 6-8 Ethnkity Asian Black Hispanic White Prevalence n % 192 3.7 118 6.1 188 11.2 1,046 11.4 Minnesota Asian 36 13.9 Suburban/Urban Black 201 4.0 Murray Hispanic 24 45.8 Native American 38 18.4 White 1,602 19.6 New York Asian 119 2.5 New York City Black 205 0.5 Grade 6 Hispanic 510 1.0 White 501 1.2 New York Asian 23 4.3 Suburban Black 47 2.1 Grade 7 Hispanic 39 2.6 Native American 26 3.8 White 1,796 3.3 Oregon Asian 38 5.3 Suburban/Rural Black 33 15.2 Grades 6-11 Hispanic 61 16.4 Native American 120 23.3 White 3.162 14.2 Oregon Asian 71 2.8 Suburban Black 231 3.9 Grades 6-9 Hispanic 26 0.0 Native American 48 12.5 White 1,847 7.6 Southeastern uIlited states 10 SMSA's Black 258 3.9 White 652 14.0 Washington Rural Grades 4-8 Asian Black Hispanic Native American White 148 6.1 119 1.7 111 9.0 179 30.7 1,434 9.4 23 both cross-sectional and longitudinal data. Hunter and her colleagues assessed tobacco use by children in Bogalusa, Louisiana, in 1976-77 and again in 1981-82 (az). The use of both snuff and chewing tobacco in- creased over time within age categories, within age cohorts, and across age categories (table 12). A decrease in use was observed in the oldest age category, 16-17 years old, but has not been seen in other locales (tables 12 and 13). The decrease may reflect agerelated changes in nor- mative behavior particular to that ares or a cohort effect. Peer and family members are found consistently to be important in- fluences on smokeless tobacco use by children and adolescents. Young users of smokeless tobacco have more friends who also use smokeless tobacco (343fQ9,&~ and may themselves identify friends' encourage ment as a reason for use (35,,44). Users of smokeless tobacco are also more likely to have family members who themselves use smokeless tobacco (34,3&$5) and encounter less parental disapproval of the prac- tice (31,359. In a special National Program Inspection study prepared by the Of- fice of the Inspector General of the Department of Health and Human Services, young current and former users of smokeless tobacco were interviewed in depth (So). `AVO hundred and ninety students in junior and senior high schools from 16 States volunteered to participate. AU had used smokeless tobacco on a weekly or daily basis. While this study was not designed to provide prevalence estimates, it provides useful in- formation about the attitudes and practices of some adolescent smoke less tobacco users. Over 90 percent of these respondents used snuff exclusively, and over 55 percent indicated that they would have strong cravings if they tried to quit. On the average, this group reported first trying snuff at age 10 and beginning regular use by age 12. Fifty percent cited pressure from friends as their primary reason for initiating use, but continued use was most often attributed to enjoyment of taste (64 percent) and habit strength ("being hooked," 37 percent). Over 85 percent thought that dipping and chewing can be harmful to health, but less than 55 percent considered regular use to present a moderate or severe risk. CONCLUSIONS 1. Recent national data indicate that over 12 million persons used some form of smokeless tobacco (chewing tobacco and snuff) in 1985 and that approximately 6 million used smokeless tobacco weekly or more often. Use is increasing, particularly among young males. 2. The highest rates of use are seen among teenage and young adult males. A recent national survey indicates that 16 percent of males between 12 and 25 years of age have used some form of 24 smokeless tobacco within the past year and that from onethird to onehalf of these used smokeless tobacco at least once a week. Use by females of all ages is consistently less than that of males; about 2 percent have used smokeless tobacco in the last year. 3. State and local studies corroborate the national survey findings. The prevalence of smokeless tobacco use by youth and young adults varies widely by region, but use is not limited to a single region. In several parts of the country, as many as 25 to 35 per- cent of adolescent males have indicated current use of smokeless tobacco. RESEARCH NEEDS More systematic and detailed national and local surveys on smoke less tobacco should be conducted.* National probability sample surveys need to be supplemented with surveys of suspected "hot spots" to detect the extent of high-risk areas in the country and the prevalence of use in these areas. Standard&d methods are essential to facilitate appropriate compari- sons among data. The current state of assessment is similar to the early days of research on cigarette smoking before standardized formats for assessment of prevalence and quantification of dosage became available. Accurate and reproducible dosage measurement for smokeless tobacco products is needed. Standardization may prove more difficult than for cigarette smoking because of the multiplicity of product forms. Specific items that require standardization include the following o Collection of data separately for snuff and chewing tobacco. o Definition of user classified according to the frequency of use. `Lb date, little attention has been given to finer distinctions of use, in- cluding quantity used, the appropriate unit of measurement, and time that the product is allowed to remain in the mouth o Description of use. Data need to be gathered on patterns of use as well as the relationship of use to cigarette smoking. o Reporting of age of initiation and duration of use. o Definition of quit attempts and a quitter. o Natural history of smokeless tobacco use and its relationship to other substance use, including other forms of tobacco, particularly cigarettes. o Surveys of adequate sizes to permit stratification of the samples by relevant variabies such as gender, age, ethnicity, socioeconomic status, cigarette smoking status, and various behavioral factors such as attitudes and knowledge, peer pressure, and academic status. * The 1986 OSH Adult Use of Tobacco Survey will address many of the items listed below. 25 REFERENCES (1) Christen, A.G., Swanson, B.B., Glover, E.D., and Henderson, A.H. Smokeless tobacco: The folkIore and social history of snuffing, sneez- ing, dipping, and chewing. J. Am. Dent. Assoc. 105: 821-829,1982. (2) Schuman, L.M. Patterns of smoking behavior. In: M.E. Jarvick, J.W. Cullen, E.R. Critz, T.M. Vogt, and L.J. West teds.). Research on smok- ing behavior (NIDA Research Monograph 17). U.S. Government Print- ing Office (Stock No. 017-024-00694-7), 1977. (3) Shelton, A. Smokeless sales continue to climb. Tobacco Reports, p. 42. August 1982. (4) Maxwell, J.C., Jr. Chewing, snuff is growth segment. Tobacco Reports, p. 31, September 1980. (5) Winn, D.M. Tobacco chewing and snuff dipping: An association with human cancer. In: I.K. O'Neil, R.C. Borstel, C.T. Miller, J. Long, and H. Bartsch (eds.). N-Nitroso Compounds Occurrence, Biological Effects and Relevancy to Human Cancers. IARC Scientific Publication No. 57, Oxford University Press, 1985. (6) Rosenfeld, L., and Cahaway, J. Snuff dipper's cancer. Am. J. Surg. 106: 840-844, 1963. (7) Agricultural Market Service. Tobacco in the United States. U.S. Department of Agriculture (Miscellaneous Publication No. 867). 1979. (8) Davis, D.L. Smokeless tobacco products and their production in the United States. Presented at the NIH Consensus Development Confer- ence on the Health Implications of Smokeless Tobacco Use, Bethesda, Maryland, January 13-15, 1986, and personal correspondence. (9) Bantle, L.F. Smokeless tobacco-A trend to watch. Ihbak J. Int. 4: 344-346, 1980. (10) Rizio, D. Smokeless tobacco. Thbak J. Int. 2: 183-184, 1984. (ll/ U.S. Department of Agriculture, Economic Research Service. Tobacco: Outlook and Situation Report. Washington, D.C., 1985. (12) U.S. Department of Agriculture, Agricultural Marketing Service. Annual report on tobacco statistics, 1973. (Statistical Bulletin No. 528). Washington, D.C., 1974. (13) U.S. Department of Agriculture, Agricultural Marketing Service. Annual report on tobacco statistics, 1976. (Statistical Bulletin No. 570). Washington, D.C., 1977. (14) U.S. Department of Agriculture, Agricultural Marketing Service. Annual report on tobacco statistics, 1981. (Statistical Bulletin No. 685). Washington, D.C., 1982. (15) U.S. Department of Agriculture, Agricultural Marketing Service. Tobacco stocks, as of January 1, 1983. Washington, D.C., 1983. (16) U.S. Department of Agriculture, Agricultural Marketing Service. Tobacco stocks, as of January 1, 1984. Washington, D.C., 1984. (17) U.S. Department of Agriculture, Agricultural Marketing Service. Tobacco stocks, as of January 1, 1986. Washington, D.C., 1986. 26 (18) National Clearinghouse for Smoking and Health. Use of tobacco: Prac- tices, attitudes, knowledge, and beliefs. U.S. Fail 1964 and Spring 1966, July 1967. (19) U.S. Department of Health, Education, and Welfare. Smoking and health-A report to the Surgeon General. Office on Smoking and Health (DHEW Publication No. PHS 79-50066). 1979. (20) National Clearinghouse to3 Smoking and Health. Adult use of tobacco, 1975. U.S. Department of Health, Education, and Welfare, Public Health Service, 1976. (21) National Center for Health Statistics. National Health Interview Survey, 1970 (unpublished). (22) Simmons Market Research Bureau, Inc. Study of Media and Markets, 1980-1985. (23) Simmons Market Research Bureau, Inc. Simmons National College Study, 1983 and 1985. (24) Office on Smoking and Health. Current Population Survey, 1985 (unpublished). (25) Rouse, B.A. National prevalence of smokeless tobacco use. Presented at the NIH Consensus Development Conference on the Health ImpIica- tions of Smokeless Tobacco Use, Bethesda, Maryland, January 13-15, 1986. (26) U.S. Department of Commerce, Bureau of the Census. (CP Series P25, No. 922), 1982. (27) Hartge, P., Hoover, R., and Kantor, A. Bladder cancer risk and pipes, cigars, and smokeless tobacco. Cancer 55: 901-906, 1985. (2%) Glover. E.D., O'Brien, K., and Holbert, D. Prevalence of smokeless tobacco use in Pitt County, North Carolina. Int. J. Addict. (in press). (29) Gritz, E.R., Ksir, C.. and McCarthy, W.J. Smokeless tobacco use in the United States: Past and future trends. Ann. Behav. Med. 7: 24-27,1985. (30) Glover, E.D., Johnson, R., Laflin, M., and Christen, A. Smokeless tobacco trends in the United States. World Smoking and Health (in press). (31) Marty, P.J., McDermott, R.J., Young, M., and Guyton, R. Prevalence and psychosocial correlates of dipping and chewing in a group of mraI high school students. Health Educ. (in press). (32) Marty, P.J., McDermott, R.J., and Wihiams, T. Patterns of smokeless tobacco use in a population of high school students. Am. J. Public Health 76: 190-192,1986. CW Newman, I.M.. and Duryea. E.J. Adolescent cigarette smoking and tobacco chewing in Nebraska. Nebr. Med. J. 66: 243-244, 1981. (34) Bonaguro, J.A., Pugy, M., and Bonaguro, E.W. Multivariate analysis of smokeless tobacco use by adolescents in grades four through twelve. Health Educ. (in press). (35) Lichtenstein, E., Severson, H.H., Friedman, L.S., and Ary, D.V. Chew- ing tobacco use by adolescents: Prevalence and relation to cigarette smoking. Addict. Behav. (in press). 27 (36) Severson, H., Lichtenstein, E., and Gallison. C. A pinch or a pouch in- stead of a puff? Implications of chewing tobacco for addictive pro- cesses. Bulletin of Psychologists in Addictive Behaviors 4: 85-92,1985. 137) Jones, R.B. Smokeless tobacco: A challenge for the 80's. Journal of the Wisconsin Dental Association 10: 71'7-721, 1985. (38) Chassin. L., Presson, C.C., and Sherman, S.J. Stepping backward in order to step forward: An acquisition oriented approach to primary prevention. J. Consult. Clin. PsychoL (in press). (39) Chassin, L., Presson. C.C., Sherman, S.J., McLaughlin, L., and Gioia, D. Psychological correlates of adolescent smokeless tobacco use. Addict. Behav. (in press). (40) Greer, R.O., and Poulson, T.C. Oral tissue alterations associated with the use of smokeless tobacco by teenagers, I. Clinical findings. Oral Surg. 56: 275-284, 1983. (41) Paulson, T.C., Lindenmuth, J.E., and Greer, R.O. A comparison of the use of smokeless tobacco in rural and urban teenagers. CA 34: 248-261, 1984. (42) Hunter, S.M., Croft, J.B., Burke, G.L., Parker, F.C., Webber, L.S., and Berenson, G.S. Longitudinal patterns of cigarette smoking and smoke less tobacco use in youth. Am. J. Public Health 76: 193-195,1986. (43) Guggenheirner, J., Zullo, T.G., Krupee, D.C., and Verbin, R.S. Chang- ing trends of tobacco use in a teenage population in western Pennsyl- vania. Am J. Public Health 76: 196-197, 1986. (44) Schaefer, S.D., Henderson, A.H., Glover, E.D., and Christen, A.G. Pat- terns of use and incidence of smokeless tobacco consumption in school- age children. Arch. Otolaryngol. (in press). (4.5) Young, M., and Williamson, D. Correlates of use and expected use of smokeless tobacco among kindergarten children. Psychol. Rep. 56: 63-66,1985. (46) Boyd, G.M.. et al. Use of smokeless tobacco among children and adolescents in the United States. Prev. Med. (in press). (47) Evans, R.I., Hansen, W.G., and Mittehnark. M.B. Increasing the valid- ity of self-reports of smoking behavior in children. J. Appl. Psychol. 62: 521-523. 1977. (48J Murray, D.M.. O'Connell, C.M., Schmiel. L.A., and Perry, C.P. The validity of smoking self-reports by adolescents: A reexamination of the logic and pipeline procedure. Addict. Behav. (in press). (49) Bauman, K.E., and Dent, C.W. Influence of an objective measure on self-reports of behavior. J. Appl. Psychol. 67: 623-638, 1982. (50) Office of the Inspector General. Youth use of smokeless tobacco: More than a pinch of trouble. U.S. Department of Health and Human Ser- vices, January 1986. 28 Chapter 2. CARCINOGENESIS ASSOCIATED WTTH SMOKELESS TOBACCO USE CONTENTS Introduction.. ................................... ..3 3 Epidemiologic Studies and Case Reports of Oral CancerinReIationtoSmokelessTbbaccoUse ............. .33 DataFromNorthAmericaandEurope .............. .33 DataFromAsia ................................ .42 summary......................................4 4 References ................................... ..4 4 Epidemiologic Studies of Other Cancers inRelationtoSmokeless'I%accoUse ................... .47 NasalCancer.................................... 8 EsophagealCancer............................... 8 LaryngealCancer................................ 0 StomachCancer ............................... ..5 1 Urinary'It-actCancer ............................ .52 Other Cancers .................................. .54 summary.. .................................. ..5 5 References ................................... ..5 6 Chemical Constituents, Including Carcinogens, of Smokeless Tobacco ............................... .58 Chemical Composition of Smokeless Tobacco .......... .58 Carcinogens in Smokeless `Ibbacco .................. .58 Summary......................................6 5 References ................................... ..6 7 Metabolism of Constituents of Smokeless Tobacco .... .70 Metabolism of NNK ........................ .71 Metabolism of NNN ........................ .73 Metabolism of NMOR ...................... .74 summary ................................ .75 References ............................... .75 Experimental Studies Involving Exposing Laboratory Animals to Smokeless `Ibbacco or Its Constituents ......... .79 Bioassays With Chewing Tobacco .................. .79 Bioassays With Snuff. ........................... .83 Bioassays With Constituents of Smokeless Tobacco .... .86 Mutagenicity Assays and Other Shoti'Ihrrn lb&s ...... .88 Summary.. .................................. ..8 9 References ................................... ..8 9 cOnclusions........................................92 ResearchNeeds.....................................93 31 This chapter presents the results of a systematic review of the world's medical literature describing experimental and human evidence perti- nent to the evaluation of smokeless tobacco as a potential cause of cancer. Five categories of research relevant to assessing the role of smokeless tobacco in cancer causation were defined: 1. Epidemiologic studies and case reports of oral cancer in relation to smokeless tobacco use. 2. Epidemiologic studies of other cancers in relation to smokeless toba~0 use. 3. Chemical constituents of smokeless tobacco. 4. Metabolism of constituents of smokeless tobacco. 5. Experimental studies involving exposing laboratory animals to smokeless tobacco or its constituents. consensus summari es of the literature in each of these categories were prepared and form the basis of this report. In addition, recommen- dations for future i-search to clarify suggestive findings or fi.lI gaps in knowledge are made. EPIDEMIOLOGIC STUDIES AND CASE REPORTS OF ORAL CANCER IN RELATION TO SMOKELESS TOBACCO USE Because smokeless tobacco products used in different regions of the world vary considerably in composition and usage patterns, this section will consider North American and European data separately from Asian data. Citations to the literature from India and other Asian coun- tries where quids containing tobacco and other ingredients are com- monly used orally focus on articles that attempt to distinguish tobacco from other ingredients in the quids as possible determinants of cancer risk. Data From North America and Europe Although about a dozen informative epidemiologic studies of smoke less tobacco use and oral cancer in North America or Europe have been reported, only a few were specifically designed to examine this relation. There are two major reasons for the relative paucity of studies. Apart from the recent increased prevalence in use of smokeless tobacco, the habit has not been widely practiced in America during this century, ex- cept in localized areas such as parts of the rural South (1,2). Further- more, cancer of the mouth is uncommon in the Western Hemisphere, 33 exacerbating the difficulty of conducting epidemiologic investigations, particularly cohort studies, into the relation between smokeless tobacco and oral cancer. The ageadjusted incidence rate for cancers of the buc- cal cavity and pharynx in the United States is approximately 11 cases per 100,000 population per year, with these tumors accounting for about 3 percent of all cancer deaths (3). Nevertheless, sufficient informa- tion is available to evaluate whether the use of smokeless tobacco increases the risk of oral cancer. case studies In their review of 566 oral cancer patients treated in two hospitals in Nashville, Rosenfeld and Callaway (4,5) noted that the proportion of women (61 percent) with buccal and gingival carcinoma was higher than the proportion of men (36 percent). Approximately 90 percent of women with buccal and gingival carcinoma used snuff for 30 to 60 years; in con- trast, 22 percent of women with cancers in other oral cavity subsites used snuff. Many of these women began practicing "snuff dipping," namely, the placement of tobacco snuff in the gingivobuccal sulcus, be tween the ages of 10 to 20 years. These reports are typical of numerous and sometimes large series of cases from the South, which reported that high percentages of patients with gingivobuccal cancers were snuff dip- pers or tobacco chewers (613). The articles describing these case series generally did not use comparison (control) groups, but the authors con- sistently commented on an apparently high prevalence of the use of snuff by the cancer patients. Clinicians also noted that the usual male predominance for epidermoid carcinomas of the oral cavity diminished or disappeared for the subgroup of gingivobuccal carcinomas occurr@ in geographic areas where there was relatively common use of snuff and chewing tobacco. Ahblom reported in the 1930's on a possible association between smokeless tobacco and cancer in Sweden (14). Among male patients with cancers of various sites seen at the Ftadiumhemmet (Stockholm), the use of snuff or chewing tobacco was reported in 70 percent with buc- cal, gingival, and "mandibuIar" cancers as compared to 26 to 37 percent with cancers in other oral subsites, the larynx, pharynx, and esophagus. Ax611 et al. reviewed medical records of male patients with squamous cell carcinoma in the oral cavity diagnosed between 1962 and 1971 and recorded in the Register of the Swedish Board of Health and Welfare (Is). The authors were only able to determine a history for the pattern of use of snuff in 25 percent of eligible patients but commented that two thirds of patients who were verified snuff users had oral cancers in regions where the snuff was generally placed. Reports of a single or a few cases, usually among male tobacco chewers, in the Northern United States and Canada also described buccal carcinomas that were often located precisely in the area where the tobacco was retained in the mouth (1619) 34 In the early 1940's, Friedell and Rosenthal associated the use of snuff or chewing tobacco with an exophytic, verrucous type of squamous car- cinoma of the oral cavity (16). Ackerman described in detail the morpho logic and clinical features of verrucous carcinoma of the oral cavity (20). Where the lesions originated in the buccal mucosa, a history of chronic use of chewing tobacco was elicited in 60 percent of the patients. The morphologic description was that of a well-differentiated, locally inva- sive, papillary squamous carcinoma, often in association with leuko plakia. In more than half of these patients, there was poor oral hygiene and carious and missing teeth. In summary, clinical and pathological reports published during the past four decades in the United States and elsewhere have commented on the use of smokeless tobacco by oral cancer patients and have described the entity known as snuffdipper's carcinoma (4,7,11), providing the basis for the hypothesis that the prolonged use of snuff or chewing tobacco is associated with an increased risk of low-grade, verrucal or squamous cell carcinoma of the buccal mucosa and gingivobuccal s&us. case control studies Most of the epidemiologic evidence comes from several case-control studies of oral cancer. The low prevalence of smokeless tobacco use in most North American populations contributes to a low statistical effi- ciency in most of these studies. Good information has been obtained, however, from studies that were either very large, conducted in an area of high prevalence of smokeless tobacco use, or analyzed according to site within the oral cavity (since the tissue affected by snuff use appears to be highly localized). One study, by Winn et al., with these characteris- tics consequently provides the most informative body of data on the carcinogenicity of smokeless tobacco in North America (21). The major concern for validity in the epidemiologic studies of smoke less tobacco and oral cancer is uncontrolled confounding. A smalI num- ber of subjects in crucial categories prevented efficient adjustment for confounding by stratification in many of these studies. Many of the studies were conducted before the advent of sophisticated epidemic logic analyses and make no attempt to control confounding. The two primary confounding factors of concern are alcohol consumption and smoking (22). Alcohol consumption is a strong risk factor for oral can- cer. It is not clear on a priori grounds, however, to what extent alcohol consumption would be correlated with smokeless tobacco use. The rela- tion between smoking, also a strong risk factor for oral cancer (2), and smokeless tobacco use may be complex. Users of smokeless tobacco may be more likely to have been smokers at some time. On the other hand, heavy users of smokeless tobacco typically cannot be heavy users of cigarettes, so that smoking is p resumably negatively correlated with smokeless tobacco use. Failure to control confounding by smoking would therefore lead to underestimates of the effect of smokeless tobacco. 35 TABLE l.-Smokeless Tobacco and Mouth Cancer, Case-Control Data From Moore et al. (23,24) Smokeless Tobacco Mouth Cancer Cases controls Users 26 12 Nonusers 14 26 Totals 40 38 Crude RR = 4.0 95%.Confidence Interval: 1.6-10 Chronologically, the first casecontrol study of smokeless tobacco was conducted by Moore et al. in Minnesota (23,24). Patients at the University of Minnesota `lhrnor Clinic with a diagnosis of cancer of the mouth were interviewed about tobacco use as part of a general inter- view procedure for clinic patients. Surgical outpatients who received the same interviews served as controls. Prom the data that were reported by these authors, one can calculate a crude relative risk estimate for mouth cancer among smokeless tobacco users of 4.0 with a 95percent confidence interval of 1.6-10 (table 1). An oddity was an ap- parent lack of effect for other forms of tobacco use. A partial explana- tion might be negative confounding between smokeless and smoked tobacco; indeed, 26 of the 40 cases of mouth cancer chewed tobacco. Still, the extent of disparity in crude effect estimates for smokeless tobacco (relative risk estimate 4.0) and smoked tobacco (all relative risk estimates < 1.0) is surprising. Wynder et al. reported on a case-control study of squamous cell cancers of the upper alimentary and respiratory tract that was con- ducted at Sweden's Radiumhemmet in 1952-55, including 33 tongue cancer patients, 14 lip cancer patients, 19 gingival cancer patients, and 8 patients with cancer of the buccal mucosa, among others (25). Con- trols were patients with cancers of the skin, head, and neck other than squamous cell carcinoma, stomach cancer, lymphoma, salivary-gland tumors, leukemia, sarcoma, cancers of the colon and rectum, and cancers of the female genital tract. A variety of risk factors was exam- ined, including the use of chewing tobacco. The authors state that the data suggested that an increased risk is associated with the duration of chewing tobacco for cancers of the gingiva and oral cavity but not for cancers of the tongue, lip, hypopharynx, esophagus, or larynx, but the data as presented do not permit an estimation of risk. In addition, data were not adjusted for other potential confounders, including cigarette smoking. Wynder and colleagues also reported in 1957 data from a similar hospital-based casecontrol study of mouth cancer conducted in New York (26). ?bbacco chewing was found to be more common among men with oral cavity cancers than among controls; but it was noted that almost all of these patients also drank alcoholic beverages and smoked and no further analyses were attempted. 36 TABLE 2.-Smokeless Tobacco and Mouth Cancer, CaseControl Data From Peacock et al. (27) Age Smokeless Tobacco User Nonuser Total 40-49 59-59 60-69 case controls case controls Case Controls 0 16 7 13 18 20 5 14 6 16 9 37 5 60 13 29 27 57 RR=0 RR = 1.4 RR = 3.7 RRMH = 2.0 95%.Confidence Interval: 1.0-4.2 Peacock et al. studied 56 cases of mouth cancer, including malignan- cies of the buccal mucosa, alveolar ridge, and floor of the mouth, and compared their tobacco histories with those of two control groups: 146 hospitalized controls with diagnoses other than cancer and 217 outpa- tients (27). Agespecific results using the hospitalized controls are sum- marized in table 2. The overall relative risk was estimated to be 2.0 (95percent confidence interval 1.0-4.2); the relative risk seemed to in- crease with age with an estimate of 3.7 for the 60 to 69 age group. The data were not reported in sufficient detail to control for confounding by smoking, which presumably led to underestimates of the relative risk. There was also insufficient detail reported to evaluate the relation be tween the risk of mouth cancer and the amount or duration of smokeless tobacco use. In Atlanta, patients with oral, pharynx, and larynx cancer were com- pared to three control groups having other mouth diseases, other can- cers, or no cancer (28. Among urban women, 40 percent of the cases used snuff compared to 3 percent or less of the controls (table 3). Among rural women, 75 percent dipped snuff compared to 20 percent or less among controls. Cigarette smoking was common in urban women and not specifically controlled for. Few rural female cases smoked cigarettes (7 percent) so confounding by smoking was minimal. The association between snuff dipping and oral, pharynx, and larynx cancer in women was generally evident in most age groups. Among the cases, the propor- tion of snuff dippers was highest among oral cancer patients: 53/72 were dippers compared to 2/18 pharynx and larynx cancer patients. Among men, insufficient information was provided to obtain precise epidemic logic estimates of the effect of chewing tobacco, although date from one of the bar charts presented indicate that urban cases were more likely to be users of smokeless tobacco than controls, that rural men with oral, pharynx, and larynx cancer or mouth disease were more likely to chew than controls, and that oral cancer patientu were more likely to chew 37 TABLE 3.-Estimated Relative Risks Associated With Snuff Use for Cancers of the Oral Cavity, Pharynx, and Larynx, Case.C!ontrol Data From Vogler et aL (2B), Females Only oral/ Other PlLSryIlXl Mouth Larynx Disease Other No ClUlCer Cancer Urban User Nonuser Crude Relative Risk Estimate RlUal User Nonuser Crude Relative Risk Estimate 15 1 5 4 23 56 165 373 60.8 1.7 2.8 1.0* 41 4 26 17 14 33 103 133 22.9 0.9 2.0 1.0* TABLE 4.-Smokeless Tobacco and Head and Neck Cancer by Anatomic Site, CaseControl Data From Vincent and Marchetta (29), Males Only Smokeless Tobacco Use User Nonuser Total Relative Risk Estimate 95%~Confidence Interval Control Larynx 5 2 95 21 100 23 1.8 0.3-9.8 oral All Head Phw Cavity and Neck 3 9 14 30 24 75 33 33 89 1.9 7.1 3.5 0.4-8.3 2.4-21 1.3-9.8 than the pharynx and larynx cancer cases. Among men, confounding by smoking could not be ruled out. Vincent and Marchetta reported the results of a case-control study of head and neck cancer according to anatomic site. Table 4 summarizes the findings for males (29). The oral cavity seems to be the anatomic site where the bulk of the effect is noted; only mild increases in risk were estimated for the larynx and pharynx, whereas users of smokeless tobacco were estimated to have a sevenfold greater risk for cancer of the oral cavity. These estimates are imprecise because of the small number of subjects and are uncontrolled for age and smoking. 38 TABLE 5.-Estimated Relative Risk for Cancer of the Head and Neck From Smokeless `lbbacco Use by Anatomic Site, Third National Cancer Survey (311, Males Only Relative Risk Estimate Anatomic Site Low Exposure High Exposure Gum-Mouth 5.6 3.9 Pharynx 0.6 - Lip-Tongue 0.3 1.1 LarYm 2.0 1.7 Martinez reported on a case-control study in Puerto Rico of risk fac- tors for cancers of the mouth, pharynx, and esophagus (XI). This population-based study included 400 cases of epidermoid carcinomas of those sites and 1,200 controls matched on age ( + 5 years) and sex to the cases. One control per case was drawn from the same hospital or clinic and two from the same community. There were 153 cases of mouth cancer (115 male and 38 female) and 68 cases of pharyngeal cancer (55 male and 13 female). The authors concluded that "Patients with cancer of the mouth did not often use chewing tobacco disproportionately. " However, calculation of the relative risks of mouth cancer that are asso ciated with chewing tobacco based on comparing the use of chewing tobacco only with no tobacco use suggests a strong effect for oral and pharyngeal cancer in males (data from table 13 in the paper). The esti- mated relative risks were 11.9 (95percent confidence interval 2.5-56.4) for oral cancer and 8.7 (95percent confidence interval 1.4-54.5) for pharyngeal cancer among chewers. These numbers do not include the experience of the many study subjects whose use of tobacco was mixed" (that is, those who used any combination of cigarette, cigar, and pipe smoking and chewing tobacco), and these calculations were based on unmatched data. Further evidence for the site specificity arose from a case-control analysis of multiple cancers using data from the Third National Cancer Survey (31). There were few female users of smokeless tobacco and scanty data by site within the head and neck region even for males; the findings do seem to indicate that the effect is greater for the site that is labeled gum-mouth as opposed to other head and neck sites (table 5). Browne et al. conducted interviews with 75 oral cancer patients, or (usually) their next of kin, and 150 living sex-, neighborhood-, and occupation-matched controls in the West Midlands area of the United Kingdom where oral cancer mortality rates were high and tobacco chewing was common among miners (32). Controls on average were born about 10 years earlier than the cases. The proportion of tobacco chewers was approximately the same among the 16 cases and 43 con- 39 trols who were miners, although data on this variable were missing for onefourth of the cases, and the authors apparently assumed that all cases with missing information were nonchewers. If the proportion of tobacco chewers among the cases with missing information was similar to those miners with known information, then the data would have shown a positive association between chewing tobacco and oral cancer. All of the miners with oral cancer who chewed tobacco also smoked pipes, further complicating interpretation of this study. Additional evidence that a carcinogenic effect of smokeless tobacco may be greatest at the anatomic site of exposure came from Westbrook et al. who compared the medical records of 55 female patients with cancers of the alveolar ridge or buccal mucosa who were treated at the University of Arkansas with those of 55 randomly selected female hospital controls (33). Fifty of the cases, but only one control, were snuff dippers, with the tumors among the cases typically appearing at the site where the snuff was usually placed. No reliable estimates of risk can be derived from this study because of the strong possibility that them was not comparable elicitation of exposum information for cases and controls. `Iwo large case-control studies were not reported in a way that enables a meaningful quantitative assessment of the effect of smokeless to. bacco in chewers and dippers compared to tobacco abstainers (34,35). The first study found that 10 percent, and the second 9 percent, of male oral cancer cases had ever chewed tobacco, while the corresponding fig- ure for controls was 9 percent. These studies, like many of the others cited here, were not undertaken specifically to evaluate the carcino genicity of smokeless tobacco. Although the data seem to indicate a weak relation, if any, between smokeless tobacco and cancer of the oral cavity, the findings are uncontrolled for age, race, geography, and smoking. The recent casecontrol study of Winn et al. is by far the most infor- mative study on the carcinogenicity of smokeless tobacco (21). The case series comprised 255 women with oral and pharyngeal cancer who were living in 67 counties in a high-risk (for oral cancer) region of North Carolina. ?tyo female controls were obtained for all but a few cases and were individually matched for age, race, source of ascertainment (hospital or death certificate), and county of residence. There was a four- fold increased risk of oral-pharyngeal cancer among nonsmoking white women who dipped snuff. The association could not be explained by smoking or alcoholic beverage consumption (21), denture wearing or poor dentition (36), diet (37), or mouthwash use (38). The data provided evidence for a strong relation between the duration of snuff use and risk for cancer, as well as a striking localization of the carcinogen&y to the gum and buccal mucosa (table 6). For long-term chronic users of snuff, there was nearly a fiftyfold increase in risk for cancers of the gum and buccal mucosa. Indeed, almost all of the patients with cheek and gum cancers had dipped snuff. 40 TABLE 6.--Estimated Relative R&k of tipharyngeal Cancer According to Duration of Snuff Use and Anatomic Site, Winn et al. (21) Anatomic Duration of Site Snuff use (yrb Relative Risk Estimate 95%. Confidence IUtelVal Gum and Buccal Mucosa Other Mouth and Pharynx 0 1 - 24 25 - 49 1 50 0 1 - 24 25 - 49 2 50 1.0 - 13.8 1.9 - 98 12.6 2.7 - 53 48.0 9.1 - 250 1.0 - 1.7 0.4 - 7.2 3.8 1.5 - 9.6 1.3 0.5 - 3.2 Although some of the exposure information came from interviews with next of kin, when the analysis was restricted to interviews with study subjects, the association between snuff and oral cancer was even stronger @9). Matched conditional logistic analysis yielded similar results (35). Based on calculations of attributable risk, the authors estimated that 87 percent of these cancers were due to the patients' snuff-dipping habits. The authors also provided data that demonstrated the negative confounding by tobacco smoking in the population, raising the possibility of a serious validity problem with the other studies that did not control for smoking. If the negative correla- tion between the use of smokeless and smoked tobacco holds in other populations, estimates of the carcinogenic effect of smokeless tobacco in studies without the control of smoking may be underestimates. The quantitative information that was provided by the Winn et al. study led its authors to conclude that the long-standing use of smokeless tobacco by Southern women was the principal cause of the elevated mortality from oral cancer among women in the Southern United States. Cohort Studies Few cohort studies of smokeless tobacco have been undertaken because of the rarity of both the exposure (smokeless tobacco use) and the outcome (oral cancer) of most interest. Bjelke and Schuman (40) reported on cancer mortality in cohorts of 12,945 Norwegian men and 16,930 American men and found increases in the risk of death for can- cers of the buccal cavity, pharynx, and esophagus (relative risk estimates ranged from 2.6 to 3.1(41); no further detail was given). They noted a negative association between smoking and chewing tobacco, confirming the pattern that was observed from the case-control research. In a 16.year followup of U.S. veterans, Winn et al. reported no deaths from oral or pharyngeal cancer among 951 smokeless tobacco 41 users who did not use other forms of tobacco (about 0.5 deaths were ex- pected) but a significant increase in both oral and pharyngeal cancers among smokeless tobacco users who were light smokers (42) These data, as well as those from Bjelke and Schuman (40). were reported only as abstracts in scientific journals or proceedings, with little or no detail as to the methods used, hindering interpretation of the results. Smith and colleagues followed a group of about 1,500 patients with changes in the oral mucosa to evaluate the effects of smokeless tobacco use (43,44). No oral cavity cancers were found in about 16,000 person- years of followup. Based on the results of other studies, two or three should have been detected over the study period. Smith gave little docu- mentation of the methods that were employed for followup; however, 12 percent of the original group (201 subjects) were lost without any data on outcome, and there was apparently no effort to trace them. It seems likely that persons who died and persons who developed cancer, includ- ing some with tumors of the oral cavity, may have been lost to followup. In fact, no deaths among cohort members were reported, whereas perhaps as many as 100 or more would have been expected among such a cohort of middleaged adults, making Smith's data uninterpretable. Data From Asia The highest rate-e of oral cancer among the more than 100 that are listed from population-based registries around the world that report stan- dadized cancer incidence statistics are found in India (45). In many areas of Asia, hospital statistics suggest that oral cancer is extremely common and often accounts for 25 or more percent of all cancers (4649), propor- tions that are far greater than in most areas of the United States where oral cancers typically comprise only 3 percent of all malignancies (3). It has long been thought that the chewing of quids that contain tobacco and other substances is the cause of the increased risk of oral cancer in these areas (50). The smokeless tobacco products that are commonly used include to bacco with betel leaf, areca nut, and lime mixtures (often referred to as "pan"); Khaini (powdered tobacco and slaked lime paste); mishri (pow- dered partially burnt black tobacco); nass (tobacco, ash, and cotton or sesame oil; lime is used in Iran and certain Soviet Republics); and various preparations that vary locally throughout the Southeast Asia region. The inclusion of lime, areca nut, and other ingredients in many of the smokeless tobacco-containing quids hinders the evaluation of the con- tribution of tobacco per se to the increased risk of oral tumors. From five investigations, however, relative risks of oral cancer among chewers of betel quids with versus without tobacco can be calculated. Data from these case-control studies, which were conducted in Cal- cutta, Madras, Karachi, Bombay, and several parts of India and Sri Lanka (4',51+%), reveal considerably higher risks of oral cancer for the 42 TABLE 7.-Relative Risk of Oral Canax From Betel Quid With and Without `Ibbacco (With 95Percent Confidence Limit) Study Location (References) Betel Betel Quid . No With W%zut Chewing Tobacco Tobacco Habit Remarks Calcutta, India (5054) Madras, India (5154) Karachi. Pakistan (5&w Bombay, India (53) India and SI-iLanka (47) Cases 138 46 Controls 61 70 Relative risk 4.3 1.2 estimates (3.0-6.1) 10.8-1.9) Cases 219 33 Controls 35 144 Relative risk 25 0.91 estimates 115-41) 10.4-1.6) 135 256 25 99 Smokers not included in these data. Only buccal mucosa can- cers considered. Smokers not included in these data. Only buccaI mucosa and tongue cancer cases included. Numbers reconstructed from percentages and totals. Cases Controls ReIative risk estimates Cases Controls Relative risk wtimates cases Controls Relative risk estimates 339 474 (11~~7, 238 513 120 63 (7.Ot532, 40 216 3.6 (2.4-5.2) 44 152 3.0 (2.1-4.3) 3 8 2.9 (0.6-14) 88 Smokers not included 1.690 in these data. 129 1.340 6 47 Separate analyses indicate that ele vated risks of oral cancer associated with tobacco chew- ing are found among nonsmokers as well as smokers. Smokers not included in these data. Only buccaI mucosa cancer considered. use of tobacco-containing compared to nontobacco-containing quids (table 7). The findings thus suggest that the addition of tobacco con- tributes substantially to the elevated cancer risk among chewers, although other differences between those who use versus those who do not use tobacco-containing quids could influence the differences. Smok- ing, however, is not such a difference, since most of the investigations referred to in table 7 demonstrated high relative risks of oral cancer (with excesses among tobacco chewers often exceeding tenfold com- pared to nonquid users) among chewers who did not smoke, ruling out confounding by cigarette smoking. The studies also generally found that the large majority of oral cancer patients had been tobacco chewers and suggest that the habit of quid chewing accounts for most of the oral cancers in the diverse populations studied (5.5%). 43 Summary Numerous case reports, especially in the South, have described oral cancers among smokeless tobacco users. The tumors often arose at ana- tomic locations where the tobacco was routinely placed. The number of epidemiologic investigations evaluating the relation between smokeless tobacco and oral cancer is not large, and several studies have method- ologic limitations. The pattern of increased oral cancer risk among smokeless tobacco users, however, is generally consistent across studies, with evidence of an increasing risk with increasing duration of exposure, and with excess risks tending to be greatest for those ana- tomic sites where tobacco exposures are greatest. The best designed study was drawn from a female population in the Southern United States where exposure rates are high and potentially confounding vari- ables could be taken into account. This study showed that chronic snuff users were at substantially increased risk of oral cancers and that nearly all tumors of the cheek and gum were due to snuff use. Evidence from parts of Asia, where the prevalence of smokeless tobacco use is high and oral cancer is the most common tumor, indicates a strong asso ciation between the chewing of quids and oral cancer. Users of quids that contain tobacco have much higher oral cancer rates than users of quids that do not, and the association is not confounded by cigarette smoking, raising the possib&ty that tobacco per se contributes to the elevated oral cancer risk in this part of the world. In summary, users of smokeless tobacco face a strongly increased risk of oral cancer, particu- larly for the tissues that come in contact with the tobacco. References (1) Office on Smoking and Health. Smoking and Health: A Report of the Surgeon General. U.S. Department of Health, Education, and Welfare. Washington, D.C., U.S. Government Printing Office, 1979. (2) Blot, W.J., and Fraumeni, J.F. Geographic patterns of oral cancer in the United States: Etiologic implications. J. Chron. Dis. 30: 745-757.1977. (3) Young, J.L., Percy, C.L., and Asire, A.J. Surveillance, epidemiology, and end results: Incidence and mortality data, 1973-77. NC1 Monogr. 57, 1981. (4) Rosenfeld, L., and Callaway, J. Snuff dipper's cancer. Am. J. Surg. l&C 840-844, 1963. (5) Rosenfeld, L., and Callaway, J. Squamous cell carcinoma of the oral cavity. South. Med. J. 56: 1394-1399, 1963. (6) Landy, J.L., and White, H.J. Buccogingival carcinoma of snuff dippers. Arm Surg.27: 442-447, 1961. (7) Wilkins, S.A., and Vogler, W.R. Cancer of the gingiva. Surg. Gynecol. Obstet. 10.5: 145-152, 1957. (8) Brown, R.L., Sun, J.M., Scarborough, J.E., Wilkins, S.A., and Smith, R.R. Snuff dippers intraoral cancer: Clinical characteristics and response to therapy. Cancer 18: `2-13, 1965. 44 (9) Coleman, C.C. Surgical treatment of extensive cancers of the mouth and pharynx. Ann. Surg. 161: 634-644, 1965. (10) Fonts, E.A., Greenlaw, R.H., Rush, B.F., and Rovin, S. Verrucous squamous cell carcinoma of the oral cavity. Cancer 23: 152-160, 1969. (11) HartseIIe, M.L. Oral carcinoma as related to the use of tobacco. Ala. J. Med. Sci. 14: 188-194, 1977. (12) McGuirt, W.F. Snuff dipper's carcinoma. Arch. OtoIaryngol. 109: 757-760, 1983. (13) McGuirt, W.F. Head and neck cancer in women-a changing profile. Laryngoscope 93: 106-107, 1983. (14) Ahblom, H.E. Predisposing factors for epitheliomas of the oral cavity, larynx, pharynx, and esophagus. Acta Radial. 18: 163-185, 1937 (in Swedish). (15) Ax&II, T., et al. Snuff dipping and oral cancer-a retrospective study. Tandlakartidningen 75: 2224-2226, 1978. (16) FriedeII, H.L., and Rosenthal, L.M. The etiologic role of chewing to bacco in cancer of the mouth. JAMA 116: 2130-2135, 1941. (17/ Moertel, C.G., and Foss, E.L. Multicentric carcinomas of the oral cav- ity. Surg. Gynecol. Obstet. IOf? 652654, 1958. (18) Sorger, K., and Myrden, J.A. Verrucous carcinoma of the buccaI mucosa in tobaccochewers. Can. Med. Assoc. J. 83: 1413-1417, 1960. (19) Stecker, R.H., Devine, K.D., and Harrison, E.G., Jr. Verrucose "snuff dippers" carcinoma of the oral cavity. JAMA 189: 838-840, 1964. (20) Ackerman L.V. Verrucous carcinoma of the oral cavity. Surgery 23: 670-678,1948. (21) Winn, D.M., Blot, W.J.. Shy, C.M., et aI. Snuff dipping and oral cancer among women in the Southern United States. N. Engl. J. Med. 304: 745-749, 1981. (22) Rothman, K., and Keller, E. The effect of joint exposure to alcohol and tobacco on risk of cancer of the mouth and pharynx. J. Chron. Dis. 25: 711-716, 1972. (23) Moore, G.E., Bissmger, L.L., and ProehI, E.C. Tobacco and intraoral cancer. Surg. Forum 3: 685688, 1952. (ZX#) Moore, G.E., Bissinger, L.L., and Pro&I, EC. Intraoral cancer and the use of chewing tobacco. J. Am. Geriatr. Sot. 1: 497-506,1953. (25) Wynder, E.L., H&berg, S., Jacobsen, F., and Bruss, I.J. Environmental factors in cancer of the upper alimentary tract. Cancer 10: 470-487,1957. (26) Wynder, E.L., Bruss, I.J., and Feldman R.M. A study of the etiologicaI factors in cancer of the mouth. Cancer 10: 1300-1323,1957. (27) Peacock, E.E., Greenberg, B.G., and Brawley. B.W. The effect of snuff and tobacco on the production of oral carcinoma. Am. Surg. 151: 542-550, 1960. (28 VogIer, W.R., Lloyd. J.W., and Mihnore, B.K. A retrospective study of etiologicaI factors in cancer of the mouth, pharynx, and Iarynx. Science 15: 246-258.1962. 45 1291 Vincent, R.G., and Marchetta F. The rehrtionship of the use of tobacco and alcohol to cancer of the oral cavity, pharynx, or Iarynx. Am. J. h-g. 106: 501-505, 1963. (30) Martinez, I. Factors associated with cancer of the esophagus, mouth, and pharynx in Puerto Rico. J. Natl. Cancer Inst. 42: 1069-1094,1969. (311 WiIIiams, R.R., and Harm, J.W. Association of cancer sites with to bacco and alcohol consumption and socioeconomic status of patients: Interview study from the Third National Cancer Survey. J. Natl. Cancer Inst. 58: 525-547, 1977. (32) Browne, R.M., Camsey, M.C., Waterhouse, J.A.H., and Manning, G.L. EtiologicaI factors in oral squamous ceII carcinoma. Community Dent. Oral Epidemiol. 5: 301-306, 1977. (33 Westbrook, K.C., Sven, J.Y., Hawkins, J.M., andMcKinney. D.C. Snuff dipper's carcinoma: Fact or fiction? In: H.E. Nieburg (ed.). Prevention and Detection of Cancer. New York, Marcel Dekker, 1980. pp. 1367-1371. (34) Wynder, E.L., and Stelhnan, S.D. Comparative epidemiology of tobacco-related cancers. Cancer Res. 37: 46084622, 1977. (3.5) Wynder, E.L., Kabat, G., Rosenberg, G., and Levenstein, M. Oral can- cer and mouthwash use. JNCI 70: 255-260, 1983. (3s Winn, D.M., Blot, W.J., and Fraumeni, J.F. Snuff dipping and oral cancer. N. Engl. J. Med. 305: 230-231,198l. (37) Winn, D.M., Ziegler, R.G., Pickle, L.W., GridIey, G., Blot, W.J., and Hoover, R.N. Diet in the etiology of oral and pharyngeaI cancers among women from the Southern United States. Cancer Res. 44: 1216-1222, 1984. 138) Blot, W.J., Winn, D.M., and Fraumeni, J.F. Oral cancer and mouthwash. JNCI 70: 251-253, 1983. (39) Winn, D.M. Smokeless tobacco and oral-pharynx cancer: The role of cofactors. Banbury Report (in press). (40) BjeIke, E., and Schuman. L.M. Chewing tobacco and use of snuff: ReIa- tionships to cancer of the pancreas and other sites in two prospective studies. Proceedings of the 13th International Congress on Cancer, 1982. p. 207. (41) International Agency for Research on Cancer. Tobacco habits other than smoking: Betelquid and areca-nut chewing and some related nitrosamines. IARC Monogr. EvaI. Carcinog. Risk Chem. Hum. 37: 103-104, 1985. (4.2) Winn, D.M., Wahath, J., Blot, W., and Rogot, E. Chewing tobacco and snuff in relation to cause of death in a large prospective cohort (Abstract). Am. J. Epidemiol. 116: 567, 1982. 1431 Smith, J.F., Mincer, H.A., Hopkins, K.P., and Bell, J. Snuff-dippers lesion: A cytologicaI and pathological study in a large population. Arch GtohuyngoI 92: 450-456, 1970. 1441 Smith, J.F. Snuff dippers lesion, a ten-year follow-up. Arch. OtoIaryn- gol.l01:2767-277, 1975. 46 (45) Waterhouse, J., Muir, C., Shanmugamnam, K., and Powell, J. Cancer incidence in five continents, Vol. IV. Lyon, France, International Agency for Research on Cancer, 1982. (46) Pindborg, J.J. Epidemiologic studies of oral cancer. Int. Dent. J. 27: 172-178, 1977. (47) Wahi, P.N. The epidemiology of oral and oropharyngeal cancer. Bull. WHO 38: 495-521.1968. (48) Hirayama, T. An epidemiologic study of oral and pharyngeal cancer in Central and Southeast Asia. BulL WHO 34: 41-69, 1966. (49) Paymaster, J.C. Cancer and its distribution in India. Cancer 17: 1026, 1964. (So) Orr, I.M. Oral cancer in betel nut chewers in Travancore. Lancet 2: 575-580, 1933. (51) Chandra, A. Different habits and this relation with cheek cancer. Bull. Cancer Hosp. Natl. Cancer Res. Center 1: 33, 1962. (52) Shanta, V., and Krishnamurthi, S. A study of aetiological factors in oral squamous cell carcinoma. Br. J. Cancer 13: 381, 1959. (537 Jafary, N.A., and Zaidi, S.H. Carcinoma of the oral cavity in Karachi, Pakistan: An appraisal. Trap. Doct. 6: 63, 1976. (54) Jus~awdh, D.J.. and Deshpande, V.A. Evaluation of cancer risk in tobacco chewers and smokers: An epidemiologic assessment. Cancer 28: 244-252, 1971. (55) Gupta, P.C., Pindborg, J.J., and Mehta, F.S. Comparison of carcine genicity of betel quid with and without tobacco. An epidemiological review. EcoL Dis. 1: 213-219, 1982. (56) Jayant. K.. Balakrishnan, V.. S anghvi, L.D., and Jussawalla, D.J. Quantification of the role of smoking and chewing tobacco in oral, pharyngeal, and esophageal cancers. Br. J. Cancer 35: 232-235, 1977. EPIDEMIOLOGIC STUDIES OF OTHER CANCERS IN RELATION TO SMOKELESS TOBACCO USE The epidemiologic studies reported in the preceding section that show an association between the use of smokeless tobacco and oral cancers, particularly malignancies of the cheek and gum, indicate that the topical exposure of tissues to tobacco can cause cancers at the site of the exposure. In the United States, the tissues in direct prolonged con- tact with the tobacco are generally those of the oral cavity. Smokeless tobacco may occasionally come in contact with other tissues. One case has been reported of squamous cell carcinoma that developed in the ear of an individual in Minnesota who habitually placed snuff in his ear for 42 years at the site where the neoplasm developed (II. Although but a single report, this highly unusual observation raises the possibility of a carcinogenic potential of smokeless tobacco at other anatomic sites when exposure is direct and prolonged. 47 Nasal Cancer In some areas of the world snuff is inhaled, so that tissues of the nasal cavity come in contact with the tobacco powder. The earliest report that links any form of tobacco to cancer was published over two centuries ago when what were probably nasal cancers were described in several patients in England who were heavy inhalers of snuff (2). There have been no systematic evaluations of snuff inhalation and nasal cancer in the United States, United Kingdom, or other European countries, most likely because both the sniffing habit and nasal cancer are uncommon, Sniffing snuff has been reported, however, to be a frequent habit among Bantu men, whose rates of nasal cancer have been reported to be high (31. In case-control studies of nasal sinus cancer reported in 1955,80 per- cent of patients with tumors of the maxillary antrum were prolonged and heavy snuff users, in contrast to about onethird of Bantu men with other cancers (4,s). The snuff used by the Bantu is thought to contain aloe plant ash, trace elements such as nickel and chromium, and other ingredients in addition to tobacco (6). Snuff use (presumably by inhala- tion) was reported not to account for the high rates of nasal adenocarci- noma among furniture makers in studies in England and Denmark, but evaluations of snuff itself as a risk factor were not undertaken (7,s). One casecontrol study of cancers of the nasal cavity and paranasal sinuses in the United States addressed the issue of smokeless tobacco (91. A total of 193 cases were identified in four hospitals in Virginia and North Carolina over a lo-year period. No association between sinonasal cancers and chewing tobacco was found (relative risk 0.7,95-percent con- fidence interval 0.4-1.5). However, a relative risk of 1.5 was observed for users of snuff (g&percent confidence interval 082.8). Risk was increased in snuff users for both adenocarcinomas (relative risk 3.1) and squamous cell carcinomas (relative risk 1.9) but not for other histologic types (relative risk 0.6) and was found for both sexes. The implications of the findings are not clear since the snuff used by the cases and controls was oral snuff not coming in contact with nasal tissues. Animal experiments, however, suggest that tumors distant to the site of exposme may l-ealllt from exposure to constituents of snuff (see the section on animal studies). An apparent excess of posterior nasal space tumors was reported among certain tribes in Kenya, and 6 or 12 cases interviewed were found to be chronic "liquid snuff" users (10). Multiple subsites of the respiratory tract were considered however, increasing the likelihood of a chance association No increased risk of nasopharyngeal cancer associ- ated with snuff use was noted in a casecontrol study in Singapore (11). Esophageal Cancer Other tissues that come in contact with constituents of smokeless tobacco in more dilute concentrations include the linings of the esopha- gus, larynx (supraglotic portion), and stomach. The results of studies of 48 7I!ABLE l.-Relative Risks of Ektphageal Cancer in Pemom Expoeed to Chewing `lbbacco and Snuff: Summa@ of Four CaseControl Studies controle Fht zz f3e.x % IReldw AUthtK No. Erpud No. Exjmd Risk* Wynder Chewing Any M 150 20 150 10 2.3 (4 < 1oyrs. 14 4 3.9 2 iop. 6 6 1.2 Williams chewing Level1 M 38 5.2 1,788 5.4 0.9 W or Snuff Level2 0 0 - WYgr cl=+% Any M 183 10.9 2,560 9.0 1.2 snuff Any M 4.4 2.7 1.7 Martinez t%ewingt Any M 120 2.5 360 3.6 1.2 (14) F 59 `11.9 177 7.3 2.7 *caldatedfrompubliabedrepultifuutpruvidedby~uthur. tRgtectedtonouamdtea. cancers of these three sites in relation to smokeless tobacco are inam- elusive. The studies are gtmemlly of limited power to detect small in- cxeases in risk, and many did not control for relevant, potentially con- founding variables. However, some studies of these three cancera do show au increaseinriskinrelationtotheuseofsmokeless~~.As shown in table 1, elevated relative risks of esophageal cancer up to twofold or higher were found in two hospital-based casecontrol studies in the United States involving 150 and 183 cancer patients (1213) and one in Puerto Rico (described in the previous section) with 179 casea (I#. One of the studies by Wynder and colleagues, however, found no evidence of an incmase in risk with duration of exposure, and all chewers were also smokers (12). The effect of smoking was not adjusted for in the other study (13). Another casecontrol study involving 120 black male cases of esophageal cancer was conducted in Washington, D.C. (15). Few of the cases or controls had used either chewing tobacco or snuff, suggesting that it did not contribute to the high rates of esophagealcancerobservedin~eareaFinally,datafromaprospective (cohort) study of U.S. veterans were analyzed to determine whether mortalityratesof~cdiseaseswereincreasedinusersofsmokeless tobacco (16,l. In the absence of smoking, the standardize mortality ratio for esophageal cancer was found to be 228, but this value was based on only one death In a cohort study of 12,945 Norwegian and 16,930 American men followed over 10 years, the risk of esophageal cancer was reported to be significantly increased among men who used 49 chewing tobacco or snuff, after controlling for age, residence, and smok- ing habits (17,18). Unfortunately, the results of both cohort studies have been published only as abstracts, so additional details are not available. Some evidence that the chewing of quids may increase the risk of esophageal cancer arises from studies in Southeast Asia. In a series of 237 cases of esophageal cancers in Sri Lanka, interview information from 111 revealed that 90 (81 percent) habitually used betel containing tobacco leaf (19). This percentage was considerably higher than the fre quency of betel chewing in the general population (30 percent). Betel chewing was more common among women. Esophageal cancer also was more common among women, an unusual observation since this cancer occurs more frequently among men in almost all areas of the world that report standardized cancer statistics @Q). Since few women were reported to smoke or use alcohol, the possibility of an etiologic role of chewing is increased. However, the potential effects of tobacco as op posed to other ingredients in the quids cannot be distinguished. In a case-control investigation in Bombay involving interviews with 305 esophageal cancer patients and nearly 2,000 population controls of age, sex, and religions similar to all head and neck cancer cases, a 2.5-fold in- creased risk of esophageal malignancy was observed (p < .Ol) among nonsmokers who chewed pan, a mixture usually consisting of tobacco, betel, lime, and other ingredients (21). The excess was higher, however, among those chewing quids without tobacco (relative risk 3.5) than with tobacco (relative risk 2.1). A more recent analysis (22) in Bombay based on 649 patients with esophageal cancer and 649 controls yielded similar qualitative findings, but the excess among users of pan without tobacco (relative risk 12.1) was accentuated compared to users of tobacco containing chews (relative risk 2.81. On the other hand, in an earlier case control investigation in southern India of several upper digestive tract tumors, including 93 esophageal cancers, increases in esophageal cancer risk were much greater among men who used betel with tobacco (c&u- lated relative risk 11) than without tobacco (calculated relative risk 2) (23,). The chewing of nass was not associated with esophageal cancer risk in a case-control study conducted in an area of Iran with among the world's highest rates for this cancer (24). Of 638 identified cases of esophageal cancer, interviews were completed with 344 and with 2 neighborhood controls matched to each case. The relative risk associ- ated with ever using nass was 0.9, with an upper limit of the 95-percent confidence interval of 1.5, suggesting that any major effect of nass on the origins of this cancer could be excluded. Laryngeal Cancer In a case-control analysis of the interview data from the Third Na- tional Cancer Survey (TNCS), Williams and Horm compared the prior use of smokeless tobacco products (in the aggregate) in persons with a 50 variety of individual types of cancer (including laryngeal cancer) with the history of such use in persons with the remaining cancers thought not to be related to tobacco use (ZEi,,. Prior experience with smokeless tobacco was divided into two levels of exposure. The estimates of the relative risks were controlled for age, race, and smoking. Relative risks of laryngeal cancer in men of 2.0 and 1.7 were found among individuals with low and high levels, respectively, of exposure to chewing tobacco or snuff. These estimates were not significantly different from 1.0. They are based on 106 cases, 11 with relatively low exposure and 5 with higher exposure, and 2,102 controls of which 98 had low exposure and 7 1 had high exposure. Only 13 female laryngeal cases were available for analysis in this study, which was insufficient to provide any meaningful results. A case-control study by Wynder and Stelhnan included 387 male cases of laryngeal cancer and 2,560 hospital controls (13). The percent- ages that had previously used chewing tobacco and snuff were 11.9 and 3.9, respectively, for the cases, and 9.0 and 2.7, respectively, for the con- trols. Based on these findings, crude relative risks of 1.4 for chewing tobacco and 1.5 for snuff were obtained. Neither estimate differs signifi- cantly from 1.0. No control for smoking or alcohol was done, although the authors state that cigarette smoking in users and nonusers of chew- ing tobacco was simiIar. Interviews with 560 laryngeal cancer patients and 2,000 controls from the general population of Bombay revealed significantly increased risks, compared to nonchewers, among chewers of betel without tobacco (relative risk 2.5) than with tobacco (relative risk 2.6) (21). I.ayngd cancer was noted to comprise an unusually high proportion of all cancer diagnoses in a hospital series in eastern India where pan chewing is com- mon, but no assessment of the role of tobacco was made (26) Stomach Cancer Zacho et al. noted that, in Denmark, both gastric cancer and use of chewing tobacco and snuff are directly related to age, more common in men than women, more prevalent in rural than urban areas, and in- versely related to socioeconomic status (27). On the basis of these obser- vations, they hypothesized that use of smokeless tobacco increases the risk of stomach cancer. Obviously, other differences among individuals within Denmark could also explain these findings. Weinberg et al. conducted a casecontrol study of stomach cancer in a coal mining region of Pennsylvania (28). Cases who had died of stomach cancer from 1978 through 1980 were compared with three control groups: persons who died of other cancers of the digestive system, per- sons who died of arterial sclerotic heart disease, and persons who lived in the same neighborhood as the case. All controls were matched to indi- vidual cases on age, sex, race, and location of residence. Data on the use 51 of various forms of tobacco were obtained by interviewing next&kin or (for neighborhood controls) the subjects themselves. About 16 percent of all men in the study had used chewing tobacco. This percentage did not differ significantly among the cases and the three control groups. No women in this study had chewed tobacco. This study provides some evidence to suggest that chewing tobacco does not increase the risk of gastric cancer, although a small increase in risk could have been missed due to lack of statistical power. The case-control analysis of the interview data from the TNCS found a relative risk of stomach cancer of 1.7 in men in the highest level of use of chewing tobacco and snuff, no increase in men in the lower use category, and no increase in women (W). These results are based on 120 male cases, 12 of which were users, and 82 female cases, 2 of which were users. The power of this analysis to detect a true increase in risk is ob viously low. The relative risk of 1.7 was not significantly greater than 1.0. In an abstract describing a cohort mortality study of U.S. veterans, the standardized mortality ratio for stomach cancer among non- smoking users of smokeless tobacco was 151, but no study details were provided (16). Urinary Tract Cancer Constituents of smokeless tobacco can enter the blood stream, and some are excreted in the urine. The kidney and bladder are thus poten- tially exposed to these agents but presuma bly in lower concentrations than are tissues of the upper aerodigestive tract. In a hospital-based case-control study in Seattle, Washington, patients who chewed to bacco were reported to be at nearly a fivefold increased risk of renal cancer compared to nontobacco users (29). Only 6 percent of the 88 male cases were chewers. No association between the use of smokeless to bacco products and either renal cell or renal pelvis cancer was reported in a case-control study of these tumors in England (39). Among 106 renal cell cancer case-control pairs in this study, 10 cases versus 11 con- trols had at some time used smokeless tobacco. Among 33 renal pelvis cancer-control pairs, 2 cases and 3 controls reported ever using smoke less tobacco products. In a large population-based study in Minnesota involving 495 cases and 697 controls, a nonsignificantly increased rela- tive risk of renal cell cancer of 1.7 (95~percent confidence interval 0.5-6.0) was found among snuff users after adjusting for smoking (31). There was a deficit in risk, however, associated with ever using chewing to bacco (relative risk 0.4, 95percent confidence interval 0.1-2.6). A review of eight epidemiologic investigations revealed no consistent evidence that the risk of bladder cancer is altered in users of smokeless to bacco products (table 2) (13,25,32&?). The National Bladder Cancer Study is the largest of the investigations of bladder cancer considered in this review (37). Cases for this study were selected through 10 population- 52 TABLE I.-Estimates of Relative Risks of Bladder Cancer in Persons Who Have Ever Used Chewing Tobacco and Snuff Relative Risks Years Fist Author Case Chewing (ref.) Diagnosed Sex Tobacco Both Snuff Wynder (32) 195783 Male 1.4* 0.7* Dunham et al. (33) 1958-64 Male 5.3*t 0.9*t - Female 1.1*t - 0.3*t Cole et al. (34) 196688 Both 1.1* 1.0* Williams and 1969-71 Malelevel 1 1.61 Horm (25) level 2 1.15 Female-level 1 0 level 2 1.78 Wynder and 1974-75 Males 0.9 0.7 stellman (13) Howe et al. (36) 1974-76 Males 0.9 Hartge et al. 137) 1977-78 Males 1.02 0.77t o Estimated from published report. t Based on analysis of nonsmokers only. based cancer registries in the United States. Controls were a random sample of the same population from which the cases came. Information was obtained from interviews of 2,982 cases and 5,782 controls. Analy- ses of smokeless tobacco use were restricted to the 340 cases and 1,227 controls who claimed never to have smoked cigarettes. Of these, 11 per- cent of the cases and 10 percent of the controls had ever used chewing tobacco, and 3 percent of the cases and 4 percent of the controls had ever used snuff. The relative risks of bladder cancer in users of chewing tobacco and snuff were estimated to be 1.0 (0.7-1.5) and 0.8 (0.4-1.6). respectively. Wynder et al. conducted a hospital-based study of 300 male bladder cancer cases (j72). Eleven percent of the 300 cases and 8 percent of the 300 hospital controls had ever used chewing tobacco; 2 percent of the cases and 3 percent of the controls had used snuff. The percentage of users was not significantly different in cases and controls, and no attempt was made to analyze the data further. Dunham et al. interviewed 493 bladder cancer patients and 527 hospi- talized controls in New Orleans (33). Among nonsmokers, there was an increased relative risk associated with chewing tobacco use among males but a deficit in risk associated with snuff use among females, but the numbers of cases involved were small (four males and three females). Cole et al. interviewed 470 cases from the Boston area and 500 population-based controls (34). Forty-six of the cases had used chewing 53 tobacco and three had used snuff. Based on the prior experience with smokeless tobacco in the controls (controlling for age and sex), 42.3 and 7.9 cases would have been expected to have used chewing tobacco and snuff, respectively. Some increase in the risk of bladder cancer was found in the TNCS survey, but none of the risks from this study are sig- nificantly different from 1.0 (table 1) (2.5). In addition, no evidence of a dose response is seen. In a second hospital-based casecontrol study (13) of similar design to the first (32), Wynder and St&man found that 8 percent and 1.9 percent of 586 cases had used chewing tobacco and snuff, respectively, com- pared to 9 percent and 2.7 percent of 2,560 controls who had used these two products. When analyses were restricted to nonsmokers in a con- tinuation of this study, a significant excess risk of bladder cancer was associated with snuff use among women, but only 3 of 76 cases were users (35). A population-based casecontrol study was conducted in three Cana- dian provinces by Howe et al, (3@. Controls were matched to individual cases on neighborhood, age, and sex. The ratio of male pairs discordant for the use of chewing tobacco was 29134, giving a relative risk of 0.9 (95~percent confidence interval, 0.5-1.6). This estimate was not altered by controlling for smoking. No female cases or controls gave a prior history of use of smokeless tobacco. In Denmark, 165 male and 47 female patients with cancer of the uri- nary bladder from a hospital serving a specific geographic area were interviewed, as were geographically-matched controls (3&Z?). The esti- mated relative risk associated with tobacco chewing was 2.0 (1.2-3.4) based on 39 exposed cases. In a logistic model containing variables for tobacco chewing, smoking, and other major correlates of bladder can- cer, the relative risk associated with chewing was 1.7 and statistically significantly higher than 1.0. The authors estimated that tobacco chew- ing might account for 9 percent of the bladder cancer diagnoses in the area. Although two studies did report elevated relative risks associated with smokeless tobacco use, on balance these studies provide little evi- dence to suggest that smokeless tobacco alters the risk of bladder cancer. It is possible that a small increase in risk has not been detected by the studies not reporting increases due to lack of statistical power. Other Cancers All other organs of the body are likely exposed to even lower concen- trations of products of smokeless tobacco via the blood. In a large prospective study in Norway, 16,7 13 individuals were inter- viewed to obtain information on the use of tobacco and alcohol and were followed up for development of pancreatic cancer (40). Sixty-three per- sons in the cohort developed this neoplasm during a lo-year followup. 54 After controlling for cigarette smoking and alcohol consumption, a rela- tive risk of 2.9 was observed in regular users of chewing tobacco or snuff (compared to nonusers). The 95percent confidence limits of this value include 1.0. Risk was greater in regular users than former or occasional current users, and a trend of increasing risk with amount used was of borderline statistical significance (P=.O6). The case-control analysis of the interview data from the TNCS (24) with respect to pancreas cancer is based on only 91 male cases (3 exposed to smokeless tobacco) and 85 female cases (none exposed); and although no increase in relative risk of pancreatic cancer in relation to smokeless tobacco was observed, the power of this study to detect such an increase is low. Other cancer sites were found to be related to the use of smokeless tobacco in the casecontrol analysis of the interview data from the TNCS (24). Relative risks for colon cancer at low and high levels of expo- sure were found to be 0.9 and 1.5 for men and 0.4 and 2.0 for women, respectively. Relative risks of cervical cancer in users of these two levels of exposure were 3.1 and 2.3. No studies have been conducted to con- firm or refute these findings. In view of the large numbers of possible associations investigated, these results should be considered of value only in generating hypotheses for further investigation. Summary The epidemiologic studies showing an association between the use of snuff and oral cancers indicate that topical exposure of tissues to smokeless tobacco can cause cancers at the site of the exposure. Case reports of neoplasms developing in the ear and nose of individuals who used snuff at these sites raise the possibility that direct exposure may increase the risk in locations besides the oral cavity. Other tissues that come in contact with constituents of smokeless tobacco in more dilute concentrations include the linings of the esophagus, larynx (supraglotic portion), and stomach. Results of studies of cancers of these three sites in relation to smokeless tobacco are inconclusive; many are of limited power to detect small increases in risk and did not control for relevant, potentially confounding variables. However, some studies of these three cancers do show an increase in risk in relation to the use of smoke less tobacco. Constituents of smokeless tobacco can enter the blood- stream, and some are excreted in the urine. The kidney and bladder are thus potentially exposed to these products and their metabolites but presumably in lower concentrations than are tissues of the upper aero digestive tract. Evidence suggests that the risk of bladder cancer is not altered to any large extent in users of smokeless tobacco products, but results from studies of kidney cancer are inconsistent. Information regarding the risks of other cancers in relation to smokeless tobacco use is sparse. 55 References (1) Root, H.D., Aust, J.B., and Sullivan, A. Snuff and cancer of the ear. N. Engl. J. Med. 262: 819-820, 1960. (2) Redmond, D.E. Tobacco and cancer: The first clinical report, 1761. N. Engl. J. Med. 2%: 18-23, 1970. (3) Higginson, J., and Oettle, A.G. Cancer incidence in the Bantu and Cape colored races of South Africa: Report of a cancer survey in the lYans- vaal. J. Natl. Cancer Inst. 24: 589-671. 1960. (4) Shapiro, M.P., Keen, P., Cohen, L., and de Moor, N.G. Malignant dis- ease in the lYansvaal, III. Cancer of the respiratory tract. S. Afr. Med. J. 29: 95-101, 1955. (5) Keen, P., de Moor, N.G., Shapiro, M.P., and Cohen L. The aetiology of respiratory tract cancer in the South African Bantu. Br. J. Cancer 9: 528-538, 1955. (6) Baumslag, N. Carcinoma of the maxi&q antrum and its relationship to trace metal content of snuff. Arch Environ. Health 23: l-5,1971. (7) Acheson, E.D., Hadfield E.H., and Macbeth, R.G. Carcinoma of the nasal cavity and accessory sinuses in woodworkers. Lancet 1: 311-312, 1967. (8) Anderson, H.C., Anderson, I., and Solgaard, J. Nasal cancers, symp- toms, and upper airway function in woodworkers. Br. J. Int. Med. 3p: 201-207, 1977. (9) Brinton, L.A., Blot, W.J., Beck er, J.A., et al. A case-control study of cancers of the nasal cavity and paranasal sinuses. Am. J. EpidemioL 119: 896-905, 1984. (10) HouJensen, K. On the occurrence of post nasal space tumors in Kenya. Br. J. Cancer 18: 58-68,1964. (11) Shanmugaratnam, K., and Higginson, J. Etiology of nasopharyngeal carcinoma: Origin and structure. In: C. Muir, K Shanmugaratnam (eds.). Cancer of the Nasopharynx. UICC Monogr. 1: 153-162,1967. (12) Wynder, E.L., and Bross, I.J. A study of etiological factors in cancer of the esophagus. Cancer 14: 389-413,1961. (13) Wynder, E.L., and St&mm, S.D. Comparative epidemiology of tobacco related cancers. Cancer Res. 37: 4608-4622,1977. (14) Martinez, I. Factors associated with cancer of theesophagus, mouth, and pharynx in Puerto Rico. J. Natl. Cancer Inst. 42: 1069-1094,1969. 05) Pottern, L.M., Morris, L.E., Blot, W.J., et al. Esophageal cancer among black men in Washington, D.C., 1. Alcohol, tobacco, and other risk fac- tors. JNCI 67: 777-783,198l. (lf$ Winn. D., Walrath, J.. Blot, W., and Rogot, E. Chewing tobaccoand snuff in relation to cause of death in a large prospective cohort (Abstract). Am. J. EpidemioL 116: 567, 1982. (17) Bjelke, E., and Schuman, L.M. Chewing tobacco and use of snuff: ReIa- tionships to cancer of the pancreas and other sites in two prospective studies. F'rowdngs of the 13th International Congress on Cancer, 1982, p. 207. 56 (18) International Agency for Research on Cancer. Tobacco habits other than smoking: Betel-quid and areca-nut chewing; and some related nitrosamines. IARC Monogr. EvaI. Carcinog. Risk Chem. Hum. 37: 103-104, 1985. (19) Stephen, S.J., and Uragoda, C.G. Some observations on oesophageaI carcinoma in Ceylon, including its relationship to betel chewing. Br. J. Cancer 24: 11-15, 1970. (20) Waterhouse, J., Muir, C., Shanmugaratnam, K., and Powell, J. Cancer Incidence in Five Continents, Vol. IV. Lyon, France, International Agency for Research on Cancer, 1982. (21) JussawaIIa, D.J., and Deshpande, V.A. Evaluation of cancer risk in tobacco chewers and smokers: An epidemiologic assessment. Cancer 28: 244-252, 1971. (22) JussawaIIa, D.J. OesophageaI cancer in India. J. Cancer Res. Chn. Oncol. 99: 29-33, 1981. (23) Shanta, V., and Krishnamurthi, S. Further study in aetiology of carci- nomas of the upper alimentary tract. Br. J. Cancer 17: B-23, 1963. (24) Cook-Mozaffari, P.J., Azordkegan, F., Day, N.E., Ressicaud, A., Sabai C., and Aramesh, B. OesophageaI cancer studies in the Caspian littoral of Iran: Results of a casecontrol study. Br. J. Cancer 39 293-309,1979. (25) WiIliams. R.R., and Horm, J.W. Association of cancer sites with to bacco and alcohol consumption and socioeconomic status of patients. Interview study from the Third National Cancer Survey. J. Natl. Cancer Inst. 58: 525-547, 1977. (26) Sarma, S.N. A study into the incidence and etiology of cancer of the Iarynx and adjacent parts in Assam. Indian J. Med. Res. 46 525-533, 1958. (27) Zacho, A., Nielsen, J., and Larsen, V. On the consumption of unburned tobacco in patients with cancer of the stomach. Acta Chir. Stand. 134: 272-274, 1968. (28) Weinberg, G.B., KuIIer, L.H., and St&r, P.A. A case control study of stomach cancer in a coal mining region of Pennsylvania. Cancer 56: 703-713, 1985. f29) Bennington, J.L., CampbeII, P.B., and Ferguson, B.R. Epidemiologic studies of carcinoma of the kidney, II. Association of renal adenocar- cinema with smoking. Cancer 22 821-823.1968. (30) Armstrong, B., Garrod A., and Doll R. A retrospective study of renal cancer with special reference to coffee and animal protein consumption. Br. J. Cancer 33: 127-136, 1976. (31) McLaughlin J.K., Mandel J.S., Blot, W.J., Schuman, L.M., MehI, E.S., and Fraumeni, J.F., Population-based casecontrol study of renal ceII car- cinoma. JNCI 72: 275-284, 1984. (32,) Wynder, E.L., Onderdonk, J., and Man@ N. An epidemiological inves- tigation of cancer of the bladder. Cancer 11: 1388-1406, 1963. &J Dunham, L.J., Rabson, A.S.. Stewart, H.L., Frank, A.S., and Young, J.L. Rates, interview and pathology study of cancer of the urinary bladder in New Orleans Louisiana. J. Natl. Cancer Inst. 41: 683-709, 1968. 57 (34) Cole, P., Monson, R.R., Haning, H., and Friedell, G.H. Smoking and cancer of lower urinary tract. N. Engl. J. Med. 284: 129-134, 1971. (35) Kabat, G.C.. Dieck, G.S., and Wynder, E.L. Bladder cancer in non- smokers. Cancer 57: 362-367, 1986. (36) Howe, G.R., Butch, J.D., Miller, A.B., Cook, G.M., E&eve, J., Morri- son, B., Gordon, P., Chambers, L.W., Fodor, G., and Winsor, G.M. Tobacco use, occupation, coffee, various nutrients, and bladder cancer. JNCI 64: 701-713. 1980. (37) Hartge, P., Hoover, R., and Kantor, A. Bladder cancer risk and pipes, cigars, and smokeless tobacco. Cancer 55: 901-906, 1985. (38) Mommsen, S., Aagaard, J., and Sell, A. An epidemiologic study of blad- der cancer in a predominantly rural district. Stand. J. Urol. Nepbrol. 17: 307-312, 1983. (39) Mommsen, S., and Aagaard, J. Tobacco as a risk factor for bladder cancer. Carcinogenesis 4: 335-338, 1983. (40) Heuch, I., Kvale, G., Jacobsen, B.K., and Bjelke, E. Use of alcohol, tobacco and coffee, and risk of pancreatic cancer. Br. J. Cancer 48: 6374x3,1983. CHEMICAL CONSTITUENTS, INCLUDING CARCINOGENS, OF SMOKELESS TOBACCO Chemical Composition of Smokeless Tobacco `RI date, at least 2,500 known compounds have been identified in pro cessed tobacco (1). Besides polysaccharides and protein tobacco con- tams Nicotzizna alkaloids (0.5-5.0 percent), alkanes (0.1-0.4 percent), &penes (0.1-3.0 percent), polyphenols (0.5-4.5 percent), phytosterols (0.1-2.5 percent), carboxylic acids (0.1-0.7 percent), aromatic hydra carbons, aldehydes, ketones, amines, amides, nitriles, N- and 0-hetero cyclic compounds, chlorinated organic compounds, alkali nitrates (0.2-5.0 percent), and at least 30 metal compounds (83). The most important habituating agent in tobacco is nicotine, the ma- jor representative of the alkaloids that constitute 0.5-5 percent of the leaf depending on the strain, variety, and agricultural practices that are employed during the tobacco cultivation. In total, the alkaloids are composed of 85 to 95 percent nicotine (4) and of other major alkaloids such as the secondary amines nornicotine, anatabine, and anabasme with lesser amounts of cotinine, myosmine, nicotyrine, 2,3'-dipyridyl, and N `-oxynicotine (5). Carcinogens in Smokeless Tobacco At present, three classes of carcinogzIls are known to occur in smoke less tobacco products: N-nitrosamines, polynuclear aromatic hydrocar- bons (PAH), and polonium-210 ( 210Po). Although chemical-analytical 58 FIGURE l.-N-Nitroeamhes in Smokeless `Ibbacco 1. Volatile Nitrosamines ' ' N-NO R----- R = -a-l, NDMA R = -C,H, NDEA 2. Nonvolatile Nitrosamines HO-CH,-Cl-l,, HO-CH+H, , N-NO NDEIA n N A0 NPYR 0 N JJO NPIP A0 NMOR H&-N-R R= -COOH NPRO R= -COOH NPIC LO R= -CH&OOH NPYRAC R= -CH&OOH NPIPAC R = -CH,-CH,-COOH NMPA R = -CH,CH,-CH,-CCOH NMBA 3. Tobacco-Specific Nitrosamhee NNN NAT NAB QLEc:3 @L":c.3 NNK NNAL data are lacking, some smokeless tobacco mixtures contain or are sus- pected to contain traces of cadmium and nickel compounds (6). formal- dehyde, and coumarin, all of which are known animal carcinogens (78). N-Nitroeamhea Tobacco leaves contain an abundance of amines in the form of pre teins and alkaloids. `lbba~ also contains up to 5 percent nitrates and traces of nitrite. Thus there is the potential for the formation of N-nitrosamines from the nitrate, nitrite, and amines during the process- ing of smokeless tobacco products. In tobacco, we distinguish between volatile nitrosamines, nonvolatile nitrosamines, and tobacuxqAfic nitrosamines (figure 1). With the exception of some N-nitrosamino 59 FIGURE 2.-Formation of `Ibbacco-Specific Nitrosamhes NICOTINE NORNICOTINE ANARASINE ANATABINE NITROSATION NNAL NNK NNN NAB NAT acids, the nitrosamin es in tobacco are animal carcinogens that are formed after harvesting of the tobacco during curing, fermentation, and/or aging. The N-nitrosamin o acid, N-nitrosoproline, occurs in pro- cessedfoodandcanalsobeformedinhumans by endogenous nitrosation of proline. This nitrosamino acid is not carcinogenic on the basis of pres- ently available data (912). Table 1 s ummarizes the available data for the volatile nitrosamin es in smokeless tobacco. Only one of the volatile nitrosamines, NDMA, has been found in U.S. looseleaf tobacco, but four nitrosamines have been found in American snuff. N-Nitrosomor- pholine is formed during tobacco processing or aging from morpholine, a cyclic amine that is not known to occur in uncontaminated tobacco (13,14) but originates from packing materials and/or flavor additives. Table 2 lists the presently known nonvolatile nitrosamines in smokeless tobacco. N-Nitrosod.iethanolamine (NDELA) in U.S. tobacco originates primarily from residues on tobacco leaves of the sucker-growth inhibi- tor maleic hydrazidediethanolamine (MH-30). Use of this formulation of the agricultural spray was banned in the United States in 1981, and the concentration of NDELA in smokeless tobaccos has markedly de creased since then (14,15). Figure 2 presents the formation of the tobaccospecific N-nitrosamines (TSNA) from the alkaloids. There is progressive nitrosation of the alka- loids during curing and processing and even during the shelf life of the commercial products (16). Table 3 summar&es the presently available quantitative data for four out of five TSNA's that are present in smoke less tobacco. The nitrosamines are detectable in snuff and tobacco prod- ucts from various parts of the world. Analyses of Swedish snuff brands manufactured between 1980 and 1985 have revealed a significant decrease of the levels of TSNA; such a trend has not been observed for U.S. snuff brands (14,16,17). It has been suggested that the lowering of TSNA levels in Swedish snuff brands is due to better control of the bac- terial content of the tobacco products. Reduced bacterial activity will probably reduce nitrite levels and, consequently, inhibit nitrosamine 60 TABLE l.-Volatile Nitroaminea in Smokeless `Ibbam (ppb)* Pruduct NDMA NPYR NPIP NMOR Fleference U.S. Looseleaf t Snuff ND - 380 (4) ND - 215 (26) ND - 1.2 (4) ND - 291 (16) ND (4) ND - 2.5 (4) ND - 107 (16) ND-696 (26) 1q14,1794 13y4,17,20, 29,%37 Sweden Chewing Tobacco Snuff ND - 0.6 (4) ND-60 (53) 0.9 - 3.7 (4) ND - 210 (27) ND (2) ND - 0.8 (2) 1736 ND - 0.5 (37) ND - 1.2 (53) 14,17,36 Canada Snuff 23 - 72.8 (2) 321 - 337 (2) 14 Denmark Chewing Tobacco Norway Chewing Tobacco India Chewing Tobacco U.S.S.R. Nass$ ND - 8.6 (6) 7.0 - 25.5 (6) ND (4) ND - 32.8 (6) 17.36 84.0 - 280 (2) 2.8 - 15 (2) 28-37 (2) 37 -220 (2) 17 ND - 0.56 (4) ND (4) 14 ND (4) 14 1.55 - 4.48 (4) ND (4) 1.74 - 8.82 (4) * Number m parentheses. number of samples analyzed t One sample also contained 8.6 ppb NDEA. e $ Also contained ND 69.6 NDEA /Ml. i? TABLE 2.-Nonvolatile Nitr osamines in Smokeless Tobacco (ppb)* Tobacco product NDELA NMPA NMBA NPRO NPYRAC NPIC NPIPAC Reference U.S. Looseleaf 224 - 680 (3) Snuff 160 - 6,800 1,250 - 7.420 (13) (51 Sweden Snuff 230 - 390 510 _ 4,400 (8) (12) Canada Plug Tobacco 110 (1) Snuff 1,180 - 2,720 (3) Germany Plug Tobacco 50 (2) Belgium Chewing 1,600 (1) Tobacco U.S.S.R. Nass India Chewing Tobacco 40 (4) 30 - 110 (4) 450 - 463 (2) 120 - 2.240 500 _ 50.900 ND - 2,000 (5) (13) (5) ND _ 260 890 - 29,500 100-300 (12) (12) (5) 100 (1) 8,800 - 16,600 (2) 500 - 700 (2) 100 (1) 3,300 (1) 200 (1) ND - 180 (4) 190 - 410 (4) ND - 6.100 (5) ND - 5,560 (12) 100 (1) 13J4p.M ND - 1,500 1315,34, 3Jm 100 - 200 14,15,*40 (5) 14 14 14 200 (1) 40 14 14 o Number in parentheses. number of samples analyzed TABLE 3.-`Ibbacco-Specific N-Nitrosamhes in Smokeless Tobacco (ppb)' Product NNN NNK NAT NAB Reference U.S. Looseleaf Plug lbbacco Snuff Sweden Snuff Plug Tobacco Canada Snuff Norway Snuff Denmark Snuff Chewing Tobacco Germany Plug lbbacco Snuff U.S.S.R. Nass Indii Chewing Tobacco Belgium Chewing Tobacco 620-8.200 (9) 3,400-4,300 D) 1,600-135,000 (21) 3.050-154.000 (34) 350-2,090 (3) 50,420-79,100 (2) 13,000-29,000 (2) 4.460-8,000 (3) 210-1.400 (4) 1,420-2,130 (2) 6,080-6,700 (2) 120-520 (4) 470-2,400 (5) 7,380 (1) ND-380 (4) 130-2,300 (5) loo-13,600 (21) 1,560-338,000 (21) 510-2,950 (34) ND-240 (3) 3,200-5,800 (2) 1,600-21,400 (34) 6%1,580 (3) 152.000-170,000 (2) 2,700-3,900 (2) 9,100-16,000 12) 1,350-7,030 (3) ND-210 (4) 30-40 (2) 1,500-1,540 (2) 20-130 (4) 130-230 (4) 970 (1) 2,680-6.170 300-2.80-O 1:; 330-500 (2) 3,920-4.370 (2) 32-300 (4) 300-450 (4) 130 (1) ND-140 (5) M-6,700 (12) 110-150 (19) ND-100 (3) 4,000-4,800 (2) l,OOO-2.400 (2) ND-60 (4) 30-50 (2) 8-30 (4) 30-70 (4) 14,17,41,42 43 614,16;17,384243 14.1~1738 l/17 14 17 16 17 14 16 14 14,41 38 E * Number in parentheses. number of samples analyzed. TABLE I.-JMimated Exposure of U.S. Residents to Nitmsamines* source of Exposure Nitrosamines Primary Exposlue Daily Intake Route ug/Person Beer Cosmetics Cured Meat; Cooked Bacon Scotch Whiskey Cigarette Smoking NDMA NDELA NPYR NDMA VNAt NDELA NNN NNK NAT+NAB Ingestion Dermd Absorption Ingestion Ingestion Inhalation Inhalation Inhalation Inhalation Inhalation 0.34 0.41 0.17 0.03 0.3 ii:: 2.9 > 16.2 1.2 Snuff DippingS VNA Ingestion 3.1 NDELA Ingestion 6.6 NNN Ingestion 75.0 NNK Ingestion 16.1 NAT+NAB Ingestion 73.4 > 164.5 o From the National Research Council /lgl. amended by data for snuff dipping 1131. In addition. it has been est.& lished that u NDMA and r n inhalation of the air in cars with new leather upholstery daily exposure amounts to 0.50 yg of .20 ug of NDEA 1181. t VNA. NDMA + NEMA + NDEA + NPYR 1371. 1 Bnmnemann et al. 113/; average values from the leading five U.S. finecut tobaccos used for snuff dipping in 1981; assumed daily consumption 10 g/day of snuff: VNA = NDMA + NPYR + NMOR. formation (17). NNK and NNN are powerful carcinogens in mice, rats, and hamsters, NAB is moderately carcinogenic, and NAT is inactive in rats in doses up to 9 mmolkg (table 3, page 82) 13). The daily exposure of an "average" snuff dipper to carcinogenic N-nitrosamin es exceeds by at least two orders of magnitude the esti- mated exposure of U.S. residents to nitrosamines in products other than tobacco products (table 4) (18,19). Furthermore, the concentrations of carcinogenic nitrosamines in snuff exceed very signifi~tly the per- missible limits for individual nitrosamines in consumer products (table 5). During snuff dipping or chewing of tobacco, the TSNA's are ex- tracted by the saliva. Consequently, the saliva of snuff dippers is reported to contain 5.0-420 ppb of NNN, up to 96 ppb of NNK, and 6.6-555 ppb of NAT (16). The saliva analyses of Indian tobacco chewers showed the presence of 1.2-220 ppb of NNN, 3.2-51.7 ppb of NAT, and up to 2.3 ppb of NNK (20,21). Recently, three additional TSNA's have been isolated from U.S. commercial snuff: 4-(methylnitrosamino)-l- (3-pyridyhbutanol-1 (NNAL), 4-(methylnitrosam.ino)-1-(3-pyridyl) butenel (NNO), and 4-(methytitrosamino)-4-(3-pyridyl)butanol-1 (Red NNA) (figure 3) (22). Additional amounts of TSNA's are most likely also formed by nitrosation processes that occur in the oral cavity during chewing (1422,23). 64 TABLE 5.-Permissible Limits for Individual N-Nitrosamines in Consumer Products Product Permissible Limit wb Whz) Agency Bacon (Meat) Beer Rubber Nipples of Baby Bottles 5 USDA* 5 FDA? 10 FDA$ Range of Individual Nitrosamines Present in Snuff Tobaccos ppb bdcd NNN 5,800 - 64.000 NNK 100 - 3,100 NAT 3,300 - 215,000 NAB 200 - 6,700 NDELA 160 - 6,800 Range in the leading 5 US. brands (1984-85) Range in 13 U.S. brands (1980-1985) o No "confirmable levels of mtrosammes' (441. t Regulation set for N-nitroscdimethylsminine 14.9 $ Regulation set for any mdividual volatde N-nitrosamine (461 Polynuclear Aromatic Hydrocarbons A number of naphthalenes have been identified in processed tobacco and especially in Latakia, which is flavor enriched by treatment with wood smoke (24,,25). While smoking tobaccos were found to contain 300-5,000 ppb of phenanthrene, 1 N-4.200 ppb of anthracene, 76-1,800 ppb of pyrene, 15-14,000 ppb of fluoranthene, and 8.5 ppb of benzo(a)pyrene (BaP) (26,27), analyses of British snuff in 1957 showed levels of 260 ppb of pyrene, 335 ppb of fluoranthene, and 72 ppb of BaP (28). In the five most popular snuff brands in the United States that were analyzed in 1985, BaP ranged from C 0.1 to 63 ppb (29). Polonium-210 This alphaemitting element has long been incriminated as a human carcinogen (30). The levels of 21oPo in dozens of U.S. and foreign cigar- ette tobaccos were between 0.1 and 1.0 pCi/g (31). In recent samples of the five leading U.S. snuff brands, 21oPo ranged from 0.16 to 1.22 pCi/g (29). It appears that 21oPo in tobacco leaves stems partially from certain types of fertilizers and airborne particles that are taken up by the tri- chomes (glandular hair) of the tobacco leaf (3133). Summary In processed tobacco, more than 2,550 chemical compounds have been identified. Among these are traces of known carcinogens such as 65 FIGURE 3.-`Ibbaax Specific N-Nitrwamines in Snuff U.S. Brands, 1985 Nitmmamines Relative Carchogenicity in Rats* Concentration ia Snuff We) (Dry Weight) A B NNN CP Q Co NAB cl" Q io NAT OH CH3 h-NO +++ 3.3 64 + -t 44 215 ybi 3 NO +++ + ? ? 1.1 6.7 1.8 0.3 trace$ 1.3 3.1 0.14 trace* 1.8 o + + + `Xmors with 1 mm&kg: + tumors with 9 mmollkg; (for type of tumors induced se table 4. page 381; + insignificant number of tumors with 9 mmollkg: ? not tested. t IsoM.ed amount.3 only. $ < 0.01 upig. PAH, 21OP0, and N-nitrosamines. The most prevalent organic carcine gas are the t.obawespecific N-nitrosamines that are formed from the Nicotiunu alkaloids during the processing of tobacco leaves. Their con- centrations in snuff exceed the levels of nitrosamines in other consumer products by over one hundredfold. During snuff dipping or chewing of tobacco, the nitrosation process continues within the mouth stimulated by oral bacteria. 66 References (1) Dube, M.F., and Green, CR. Methods of collection of smoke for analyti- cal purposes. Recent Advan. Tobacco Sci. 8: 42-102, 1982. (2) Wynder, E.L., and Hoffmann D. Tobacco and tobacco smoke. Studies in Experimental Carcinogenesis. New York, Academic Press, 1967, p. 730. (3) International Agency for Research on Cancer. Tobacco habits other than smoking Betel&d and areca-nut chewing and some related nitro samines. IARC Monogr. EvaI. Carcinog. Risk Chem. Hum. 37: 291,1985. (4) Sisson, V.A., and Serverson, R.F. Alkaloid composition of the Nicotianu species, 24. Tobacco Chemists Res. Co& 1984, p. 10. (5) Piade, J.C., and Hoffmann, D. Quantitative determination of aIkaIoids in tobacco by liquid chromatography. J. Liquid Chromatogr. 3: 1505-1515, 1980. (6) Baumslag, N., Keen, P., and Petering, H.G. Carcinoma of the maxillary antrum and its reiationship to trace and metal content in snuff. Arch. En- viron. Health 23: l-5, 1971. (7) Vainio, H., Hemminke, K., and Wilbur-n, M. Data on the carcinogenicity of chemicals in the IARC Monographs Programme. Carcinogenesis 6: 1663-1665, 1985. (8) Grigg, G.W. Genetic effects of coumarins. Mut. Res. 47: 161-181, 1977178. (9) Ohshima H., and Bartsch, H. Quantitative estimation of endogenous nitrosation in humans by monitoring N-nitroso proline excreted in the urine. Cancer Res. 41: 3658-3662, 1981. (10) Hoffmann D., and Brunnemann K.D. Endogenous formation of N-nitroso proline in cigarette smokers. Cancer Res. 43: 5570-5574,1983. (11) International Agency for Research on Cancer. Monograph on the evaluation of the carcinogenic risk of chemicals to humans, Vol. 17. Some N-nitroso compounds. Lyon, France, International Agency for Research on Cancer, 1978, p. 365. (12) Preussn-mm R., and Stewart B.W. N-nitroso carcinogens. In: C.E. Searle ted.). Chemical Carcinogens, Second Edition. Am. Chem. Sot. Monogr. 182: 643-828,1984. (13) Brunnemann, K.D., Scott, J.C., and Hoffmann, D. N-NitrosomorphoIine and other volatile N-nitrosamines in snuff tobacco. Carcinogenesis 3: 693496,1982. (14) Brunnemann, K.D., Genoble, L., and Hoffmann, D. N-nitrosamines in chewing tobacco: An international comparison. J. Agr. Food Chem. 33: 1178-1181, 1985. (25) Brunnemann, K.D., and Hoffmann, D. Assessment of the carcinogenic N-nitrosodiethanolamine in tobacco products and tobacco smoke. Car- cinogenesis 2: 1123-l 127, 1981. (16) Hoffmann, D., and Adams, J.D. Carcinogenic tobacco-specific N-nitrosamines in snuff and in the saliva of snuff dippers. Cancer Res. 41: 4305-4308, 1981. 67 (17) ijsterdahl B.G., and Slorach S. N-nitrosamines in snuff and chewing to bacco on the Swedish market in 1983. Food Additiv. Contamin. 1: 299-305, 1984. (18) National Research Council. In: The health effects of nitrate, nitrite and N-nitroso compounds (Ch. 7, Pt. 1). Washington, D.C., National Academic Press, 1981, p. 51. (19) Hoffmann, D., and Hecht, S.S. Nicotinederived N-nitrosamines and tobacco-related cancer. Current status and future directions. Cancer Res. 45: 935-944, 1985. (20) Nair, J., Ohshima, H., Friesen, M., Croisy, A., Bhide, S.V., and Bartsch, H. `Ibbaccospecific and betel nut-specific N-nitroso compounds. Occur- rence in saliva and urine of betel quid chewers and formation in vitro by nitrosation of betel quid. Carcinogenesis 6: 295-303, 1985. (21) Wenke, G., Rivenson, A., Brunnemann, K.D., and Hoffmann, D. For- mation of N-nitrosamines during betel quid chewing. IARC Sci. Publ. 57: 859-866, 1984. (22) Brunnemann, K.D., Chou, D.. Adams, J.D., and Hoffmann, D. On the isolation and identification of new tobaccospecific N-nitrosamines (Abstract). 39th Tobacco Chemists Res. Conf., Montreal, October 2-5, 1985. (a) Sipahimalani, A.T., Chada, M.S., Bhide, S.V., Pratap, A.I., and Nair, Y. Detection of N-nitrosamines in the saliva of habitual chewers of to bacco. Food Chem. Toxicol. 22: 261-264, 1984. (24) Schmeltz, I., Tosk, J., and Hoffmann, D. Formation and determination of naphthalenes in cigarette smoke. Anal. Chem. 48: 645-650,1976. (2.5) NicoIaus, G., and Ehnenhorst, H. Nachweis and quantitative Bestim- mung von AIkyInaphthaIinen in Latakia-?gbak. Beitr. lbbakforsch. 11: 133-140, 1982. (26J Campbell, J.M., and Lindsey, A.J. PolycycIic hydrocarbons extracted from tobacco: The effect upon total quantities found in smoke. Br. J. Cancer 10: 649-652, 1956. (27) Onishi, I., Nagasawa, M., Tomita, H., and Fukusumi, T. Studies on the essential oil of tobacco leaves, Part XVI. Neutral fraction (3). PolycycIic aromatic hydrocarbons of Burley tobacco leaf. Bull. Agr. Chem. Sot. Japan 22: 17-20.1958. (28) Campbell, J.M., and Lindsey, A.J. PolycycIic aromatic hydrocarbons in snuff. Chem. Ind. London, 951,1957. (29) Hoffmann, D., Harley, N.H., Fisenne, I., Adams, J.D., and Brunnemann, K.D. Carcinogenic agents in snuff. JNCI (in press). (30) Lundin, F.E., Jr., Wagoner, J.K., and Archer, V.E. Radon daughter exposure and respiratory cancer. Quantitative and temporal aspects. U.S. Dept. of Health, Education, and Welfare; Joint NIOSH/NIEHS Monogr. 1, 1971. (31) Harley, N.H., Cohen, B.S., and `Iso, T.C. Polonium-210: A questionable risk factor in smoking-related carcinogenesis. Banbury Report 3: 93-104. 1980. 68 (32) Martell, E.A. Radioactivity of tobacco trichromes and insoluble cigar- ette smoke particles. Nature 249: 215-217, 1974. (33) `l%o, T.C., Harley, N.H., and Alexander, L.T. Source of lead-t&in and polonium-t&in. Science 153: BBO-882,1966. (34) Brunnemann. K.D., Scott, J.C., and Hoffmann, D. N-Nitrosoproline, an indicator for N-nitrosation of amines in processed tobacco. J. Agr. Food Chem. 31: 905-909, 1983. (35) Palladino, G., Adams, J.D., B runnemann, K.D., Haley, N.J., and Hoffmann, D. Snuff-dipping in college students: A clinical profile. Mili- tary Med. (in press). (36) &terdahl, B.G., and Slorach, S.A. Volatile N-nitrosamines in snuff and chewing tobacco on the Swedish market, Food Chem. lbxicol. 21: 759-762, 1983. (37) Brunnermum, K.D., Yu, L., and Hoffmann, D. Assessment of carcino genie volatile N-nitrosamines in tobacco and in mainstream and sidestream smoke from cigarettes. Cancer Res. 37: 3218-3222, 1977. 08) Nair, J., Ohshima, H., Malaveille, C., F riesen, M., Bhide, S.V.. and Bartsch, H. N-Nitroso compounds (NOC) in saliva and urine of betel quid chewers: Studies on occurrence and formation. Carcinogenesis 6: 295-303, 1985. (39) Ohshima, H. Identification and occurrence of new N-nitrosamino acids in human urine and environmental samples. Presented at the Con- ference on Organic and Biological Chemistry of Carcinogenic and Car- cinostatic Agents Containing Nitrogen-Nitrogen Bonds, Harper's Ferry, West Virginia, May 17-21, 1985. (40) Oh&ma, H., Nair, J., Bourgade, M.C., Friesen, M., Ganeen, L, and Bar&h, H. Identification and oczaurence of two new N-nitrosammo acids in tobacco products: 3jN-nitrosoN-methyiaminoylamino)propionic acid and 4-(NnitxosoNmethylaminolbutyric acid Cancer L&t. s 153-162,1985. (41) Hoffmann, D., Hecht, S.S., Omaf, R.M., Wynder, E.L., and `lbo, T.C. Nitrosonomiwtine: Presence in tobacco, formation and carcinogenicity. IARC Sci. Pubi. 14: 307-320,1976. (42) Munson, J.W., and Abdine, H. Determination of N-nitrosonomiwtine in t.obacco by gas chromatography/mass spectroscopy. Anal. Letters 1Q 777-786, 1977. (43) Adams, J.D., Bnmnemann, K.D., and Hoffmann, D. Rapid method for the analysis of tobaccospecific N-nitrosammes by gas-liquid chroma- tography with a thermal energy analyzer. J. Chromatogr. 256: 347-351, 1983. (44) U.S. Department of Agriculture, Food Safety and Quality Service. Nitrates, nitrites and ascorbates (or isoascorbah) in bawn. Fed. Reg. & 20992-20995, May 16,1978. 1&J U.S. Food and Drug Administration. Dimethyl nitrosamine in malt beverages; availability of guide. Fed. Reg. 45: 39341-39342,198O. (4s) U.S. Food and Drug Administr h a `on. Action levels of total volatile N-nitrosammes in rubber baby bottle nipples; availability of revised wm- phance policy guide. Fed. Reg. 49 5078450790,1984. 69 Abbreviations BaP NAB NAT ND NDEA NDELA NDMA NMBA NMOR NMPA NNAL NNK NNN NNO NPIC NPIP NPIPAC NPRO NPYR NPYRAC PAH 2lOPo Red NNA TSNA Benzo(a)pyrene N `-Nitrosoanabasine N `-Nitrosoanatabine Not detected Nitrosodiethylamine Nitrosodiethanolamine Nitrosodimethylamine Nitrosomethylbutyric acid Nitrosomorpholine Nitrosomethylpropionic acid 4-(Methyhr.itrosamino)-1-(&pyridyl)-1-butanol 4-(Methyhritrosamino)-l-(3-pyridyh-1-butanone N `Nitrosonomicotine 4-(Methylnitrosamino)-l-(3-pyridyl)butenel Nitrosopipecolic acid Nitrosopiperidine Nitrosopiperidine-acetic acid Nitrosoproline Nitrosopyrrolidine Nitrosopyrrolidineacetic acid Polynuclear aromatic hydrocarbons Polonium-2 10 4-(Methyhritrosamino)-4-(3-pyridyh-1-butanol Tobaccospecific nitrosamines METABOLISM OF CONSTITUENTS OF SMOKELESS TOBACCO The tobaccospecific nitrosamines 4-(methylnitrosamino)-l-(3-pyridyl)- 1 -butanone (NNK) and N `-nitrosonornicotine (NNN) are quantitatively the major known carcinogens that are present in snuff and other types of smokeless tobacco. Molecular changes that are induced in the genetic material of tobacco chewers are most likely to arise from the metabo lism of these two nitrosamin es. Although present in similar quantities, N'nitrosoanabasine (NAB) and N'-nitrosoanatabine (NAT) are less car- cinogenic than NNK and NNN and are less likely to play an important role in the induction of oral cancer in man. Some snuff products contain considerable amounts of N-nitrosomorpholine (NMOR) and N-nitro 70 FIGURE l.-Metabolic Pathways of NNK sodiethanolamine (NDELA); the former is a potent carcinogen. The levels of benzo(ab>yrene (BaP) and 2loPo in snuff tobacco are low com- pared to those of the nitro samines (see previous section). This section will focus on the routes of metabolic activation of the compounds that are most likely to be involved in the induction of tumors that are related to snuff use-NNK, NNN, and NMOR. Metabolism of NNK The overall metabolic scheme for NNK, as determined by in uivo and in vitro studies in F-344 rats, Syrian golden hamsters, and A/J mice, is illustrated in figure 1 (l-4). A key feature of this metabolic scheme is the conversion of NNK to the alpha-hydroxy intermediate 4, which is un- stable and undergoes spontaneous conversion to the keto aldehyde 8 and, most likely, methyl diazohydroxide 9. The latter is a methylating agent that is well known for its ability to methylate DNA forming 7-methylguanine, 06-methylguanine, 4-methylthymidine, and a spec- trum of other products (5). Among these, O&methylguanine, which is generated from precursors such as N-methyhritrosourea (NMU) or N-nitrosodimethylamine, has been unequivocally shown to be able to in- duce miscoding during DNA replication, and the resulting point muta- tion is sufficient to activate proto-oncogenes (6,7). Many studies have demonstrated a correlation between 06-methylguanine persistence in rep licating tissues and the initiation of the carcinogenic process, although it is clear in other cases that additional factors are also involved (89). 71 FIGURE 2.-Scheme Linking Nicotine to Formation of the Promutagenic DNA Adduct, 06-Methylguanine TOBACCO PROCESSING METABOLIC OR PCTIVbTIOI(- [CHSN=NOtl] - 7-YETWLGUANINE CIGARETTE YETHYLOIAZO- 06-YETHYLGUANINE SYOKING HYDROXlDE IN DNA NICOTINE NNK - Recent studies have demonstrated that NNK can methylate target tissue DNA of rats; 7-methylguanine and 05methylguanine have been detected in the DNA of rat lung, nasal mucosa, and liver but not in the nontarget tissues, kidney, and esophagus (1014). These studies have also shown that, in the case of NNK, 06-methylguanine formation alone is not sufficient for tumor induction since persistent levels of 06-methyl- guanine in the lung were less than those observed upon treatment with equivalent quantities of N-nitrosodimethylamine, but the latter did not induce lung tumors (13). It is clear from these, and related studies with NNN, that DNA adducts are also formed via pyridyloxobutylation or related processes. Regardless of the mechanism, it is significant that NNK causes DNA methylation; this creates a mechanistic link between nicotine, the habituating factor in tobacco, and 08methylguanine for- mation in DNA, as illustrated in figure 2. Immunoassay methods are currently being developed to detect 06-methylguanine in the exfoliated oral cells of snuff dippers. Its presence can be inferred from the animal studies that are discussed above and by the demonstration that human tissues, including buccal mucosa, can metabolize NNK by alpha- hydroxylation (15). In this respect, it is significant that injection of Syrian golden hamsters with the methylating agent MNU, combined with irritation of the buccal mucosa, resulted in the induction of oral cavity tumors (16). The pathway of NNK metabolism leading to the alpha-hydroxy inter- mediate 3 is also considered to be important in NNK carcinogenesis. This pathway gives rise to the electrophilic diazohydroxide 7. The prop erties of this intermediate have been investigated by using a model compound, 4-(carbethoxynitrosamino)-l-(3-pyridyl)-l-butanone (CNPB). Generation of 7 from CNPB is strictly analogous to the well- known ability of NMU to generate methyl diazohydroxide. Mutagen- icity assays in S. typhimurium of CNPB have shown that it is more mutagenic than NMU (17). Chemical model studies have demonstrated that it modifies the NZ-position of deoxyguanosine (18). This adduct and other adducts that may be formed from the diazohydroxide 7 and related intermediates are likely to play an important role in tumor in- duction by NNK. Autoradiographic studies have demonstrated that radioactivity from [carbonyl-14C]NNK is firmly bound to target tissues of rats and hamsters (4,19) and to tissues of the marmoset monkey (20). 72 FIGURE 3.-Metabolic Pathways of NNN A third key feature of NNK metabolism is its rapid conversion in vivo and in cultured tissues from experimental animals and humans to its reduced form, NNAl, which has similar tumorigenic activity to that of NNK (1,34,15,21). NNAl is slowly metabolized as indicated in figure 1 and also by reconversion to NNK. Like NNK, it methyl&es DNA in vitro and in rho. While the full details of the NNK-NNAl equilibrium have not yet been elucidated, it is clear that NNAl can act as a cir- culating source of NNK metabolites. It may play an important role in tissuespecific carcinogenesis by NNK. Metabolism of NNN Metabolic pathways of NNN are illustrated in figure 3. These path- ways have been elucidated by in uiuo and in vitro studies in rats, hamsters, and mice @,3,22-29). The stable met&o& NNN-l-N-oxide (1) has tumorigenic activity somewhat less than that of NNN but is still an effective carcinogen in F-344 rats (30). Metabolism of NNN to the 2'- and 5 `-hydroxy intermediates 2 and 5 constitutes a major pathway in uiuo and in vitro in experimental animals, human liver microsomes (31), and cultured human tissues, including buccal mucosa (15). Of particular interest is the ability of two NNN target tissues, lingual mucosa and esophageal mucosa, to carry out preferential 2 `-hydroxylation of NNN @7,32). The intermediate that is formed by 2'-hydroxylation of NNN is diazohydroxide 8, which is identical to that formed by methyl hydroxy- lation of NNN (7, figure 1). As described above, this intermediate is 73 FIGURE I.-Metabolic Pathways of NMOR IO II highly mutagenic, and this or related intermediates appear to play an important role in carcinogenesis by both NNN and NNK. The inter-me diate 9 is significantly less mutagenic than 8 in S. typhimutium (33). and various lines of evidence indicate that it is less important in NNN tumorigenesis than is 8 (33,3#. Autoradiographic studies have demon- strated that radioactivity from [2'-14C]NNN is bound to tissues of mice, rats, and marmoset monkeys (ZYJ35-37). Immunoassays are currently being developed for the putative DNA adducts that are produced by 2 `hydroxylation of NNN and methyl hydroxylation of NNK; it will be important to assess the levels of these adducts in the exfoliated oral cells of snuff dippers. Their levels may relate to the susceptibility of in- dividuals to the effects of smokeless tobacco. The metabolic pathways that lead to these intermediates can be affected by alcohol consumption and dietary components &?2,38#3). Metabolism of NMOR The metabolic pathways of NMOR are illustrated in figure 4. These have been elucidated by in vitro and in uiuo studies in rats (44-47). Struc- ture activity studies bad shown that 3-hydroxylation of NMOR, leading 74 to intermediate 4, was likely to he important in NMOR carcinogenesis (48). This pathway could result in the formation of glyoxaldeoxyguano sine adducts (49); 2-hydroxylation of NMOR also occurs, giving the mutagenic product 2. The latter also forms glyoxaldeoxyguanosine adducts (50). These adducts, which are likely to have miscocling proper- ties, also should he present in the DNA of snuff dippers since human tissues are capable of metabolizing NMOR 151). Summary Persuasive evidence exists that the carcinogenic nitrosamines that are present in high quantities in snuff and other forms of smokeless to bacco are metabolized by target tissues of experimental animals and by human tissues to intermediates that can modify the genetic material of the cell. References (1) Hecht, S.S., Young, R., and Chen, C.B. Metabolism in the F-344 rat of 4-(N-methyl-N-nitrosamino)-I-(3-pyridyl)-l-butanone. a tobacco specific carcinogen. Cancer Res. 40: 4144-4150.1980. (2) Hoffmann, D.. Castonguay, A., Rivenson, A., and Hecht, S.S. Compar- ative carcinogenicity and metabolism of 4-(methyl-nitrosamino)-l- (3-pyridyl)-l-butanone and N'nitrosonornicotine in Syrian golden hamsters. Cancer Res. 41: 2386-2393, 1981. (3) Castonguay, A., Lin, D., Stoner, G.D., Radok, P., Furuya, K., Hecht, S.S., S&t, H.A.J., and Klaunig, J.E. Comparative carcinogenicity in A/J mice and metabolism by cultured mouse peripheral lung of N'-nitroeonornicotine, 4-(methyhritrosamino)-l-(3-pyridyl)-1-butanone and their analogues. Cancer Res. 43: 1223-1229,1983. (4) Castonguay, A., Tjjilve, H., and Hecht, S.S. Tissue distribution of the tobacco-specific carcinogen 4-(methylnitrosamino)-l-(3-pyridyl)-l- butanone, and its metabolism in F-344 rats. Cancer Ftes. 43: 630638, 1983. (5) Singer, B., and Grunberger, D. Molecular biology of mutagens and car- cinogens. New York, Plenum Publishing Corp., 1983, pp. 45-96. (s) Loechler, E.L.. Green, C.L., and Essigmann, J.M. In viva mutagens by O~methylguanine built into a unique site in a viral genome. Proc. Natl. Acad. Sci. USA 81: 6271-6275,1984. (7) Sukumar, S., Nofario, V., Martin-Zanca, D., and Barbacid, M. Induc- tion of mammary carcinomas in rats by nitrosomethylurea involves malignant activation of H-ras-1 locus by single point mutations. Na- ture 3iX? 658-662,1983. (8) Pegg, A.E. Methylation of the 06-position of guanine in DNA is the most likely initiating event in carcinogenesis by methylating agents. Cancer Invest. 2: 223-231. 1984. 75 (9) Singer, B. Akylation of the 06 of guanine is only one of many chemical events that may initiate carcinogenesis. Cancer Invest. 2: 223-238, 1984. (10) Castonguay, A., Tharp, R., and Hecht, S.S. Kinetics of DNA methyla- tion by the tobacco specific carcinogen 4-(methyInitrosamino)-l-(3- pyridyl)-1-butanone in the F344 rat. In: I.K. O'NeiIl, R.C. Von Borstel, C.T. Miller. J. Long, and H. Bsrtsch, (eds.). N-Nitroso Compounds: Occurrence, Biological Effects and Relevance to Human Cancer. IARC Sci. Publ. 57: 805-810, 1984. (11) Chung, F-L., Wang, M., and Hecht, S.S. Effects of dietary indoles and isothiocyana~ on N-nitror&methyIamine and 4+nethyhiitrosammo)-l- (3-pyridyl)-l-but alpha-hydroxykion and DNA methylation in rat liver. Carcinogenesis 6: 539543,1985. (12) FoiIes. P., l'kushin, N., and Castonguay, A. M easurement of Ok&hylde oxyguanosine in DNA methylated by the t.obaccoqecific carcinogen 4- (methylnitrosamino)-1-(3-pyridyl)-1-butanone using a biotin-avidin enzymelinked immunosorbent assay. Carcinogenesis 6: 989993.1985. (13) Hecht. S.S., `Ikushin, N., Castonguay, A., and Rivenson, A. Comparative tumorigenicity and DNA methylation in F344 rata by 4+nethylnitro samino)-l-(3-pyridyl)-l-butanone and N-nitrosodimethyhunine. Cancer Res. 46: 498-502,1986. (14) Belinsky, S.A., White, C.M., Boucheron, J.A., Richardson, F.C., Swen- berg, J.A., and Anderson, M.W. Accumulation of DNA adducts in hepatic and respiratory tissue foIlowing multiple administrations of the tobacco-specific carcinogen 4-(N-methyl-N-nitrosamino)-l-(3-pyridyl)l- butanone (NNK). Proc. Am. Assoc. Cancer Rea. 26: 100,1985. (15) Castonguay, A., Stoner, G.D., Schut, H.A.J., and Hecht, S.S. Metab lism of tobaccospecific N-nitro samines by cukumd human tissues. Proc. NatI. Acad. Sci. USA 80: 66946697,1983. (16) Konstantinidus, A., Smulow, J.B., and Sonnenschein, C. `Rnnorigenesis at a predetermined oral site after one intraperitoneal injection of N-nitrosoN-methylurea. Science 216: 1235-1237,1982. (17) Hecht, S.S., Lin, D., and Castonguay, A. Effects of alphadeuterium substitution on the mutagenicity of 4-(methylnitrosammo)-113-pyridylb 1-butanone (NNK). Carcinogenesis 4: 305-310,1983. (18) Hecht, S.S., Lin, D., Chuang, J., and Castonguay, A. Reactions with deoxyguanosine of 4+arbethoxynitr osamino)-l-(3-pyridyl)l-butanone, a model compound for alpha-hydroxylation of tobacco specific nitro- samines. J. Am. Chem. Sot. (in press). (19) Tj&e, H., and Castonguay, A. The in uiuo tissue distribution and in vitro target tissue metabolism of the tobaccospecific carcinogen 4- (methylnitrosamin o)-l-(3-pyridyl)1-butanone in Syrian golden hamsters. Carcinogenesis 4: 1259-1265,1983. CZWJ Castonguay, A., Tjlil,e, H., Ikushin, N., d'Argy R., and Sperber, Y. Metabolism and tissue distribution of tobaccospecific N-nitrosamines in the marmoset monkey (Cahithria jacchus). Carcinogenesis 6: 1543-1550, 1985. 76 (21) Adams, J.D., LaVoie, E.J., and Hoffmann, D. On the pharmacokinet- its of tobaccospecific N-nitro samines in Fischer rats. Carcinogenesis 6: 509511,1985. (22) Chen, C.B., Hecht, S.S., and Hoffmann, D. Metabolic alpha- hydroxylation of the tobaccospecific carcinogen N `nitrosonomicotine. Cancer Res. 38: 3639-3645, 1978. (23) Chen, C.B., Fung, P.T., and Hecht, S.S. Assay for microsomal alpha- hydroxylation of N'nitrosonomicotine and determination of the deuterium isotope effect for alpha-hydroxylation. Cancer Res. 39: 5057-5062, 1979. (24) Hecht, S.S., Chen, C.B., and Hoffmann, D. Metabolic beta-hydroxyla- tion and N-oxidation of N'nitrosonomicotine. J. Med. Chem. 23: 1175-1178, 1980. (25) McCoy, G.D., Chen. C.B., and Hecht, S.S. Influence of mixed function oxidase inducers on the in vitro metabolism of N'nitrosonomicotine by rat and hamster liver microsomes. Drug. Metab. Disp. 9: 168-169,1981. (26) Hecht, S.S., Lin, D., and Chen, C.B. Comprehensive analysis of urinary metabolites of N'-nitrosonomicotine. Carcinogenesis 2: 833-838, 1981. (27) Hecht, S.S., Reiss, B., Lin, D., and Williams, G.M. Metabolism of N'nitrosonomicotine by cultured rat esophagus. Carcinogenesis 3: 453-456,1982. (28) Hecht, S.S., and Young, R. Regiospecificity in the metabolism of the homologous cyclic nitro samines, N'nitrosonornicotine and N'-nitroso anabasine. Carcinogenesis 3: 1195-1199, 1982. (29) Brittebo, E.B., Castonguay, A., Furuya, K., and Hecht, S.S. Metabo- lism of tobacccqwcific nitrosamines by cultured rat nasal-mucosa. Cancer lb. 43: 4343-4348,1983. (30) Hecht, S.S., Young, R., and Maeura, Y. Comparative carcinogenicity in F344 rats and Syrian golden hamsters of N'nitrosonomicotine and N %ritrosonomicotinel -N-oxide. Cancer L&t. 20: 333-340,1983. (31) Hecht, S.S., Chen, C.B., McCoy, G.D., Hoffmann, D., and Domellof, L. Alpha-hydroxylation of N-nitrosopyrrolidine and N'-nitrosonomicotine by human liver microsomes. Cancer L&t. 8: 35-41, 1979. (3`2) Castonguay, A., Rivenson, A., `B-u&in, N., Reinhardt, J., Stathopoulos, S., Weiss, C.J., Reiss, B., and Hecht, S.S. Effects of chronic ethanol consumption on the metabolism and carcinogenicity of N'nitrosonomi- coke in F344 rats. Cancer Res. 44: 2285-2290, 1984. (33) He&t, S.S., and Lin, D. Comparative mutagenicity of 4&rbethoxy- nitrosamino)-4-(3-pyridyl)-butanol and 4-(carbethoxynitrosamino)-l- (3-pyridyl)-l-but, model compounds for alpha-hydroxylation of N'nitrosonornicotine. Carcinogenesis (in press). (34) Hecht, S.S., Castonguay, A., Rivenson, A., Mu, B., and Hoffman, D. Tobacco-specific nitrosamines: Carcinogenicity, metabolism, and possi- ble role in human cancer. J. Environ. Health Sci. 1: l-54, 1983. (35) Brittebo, E.B., and Tjiilve H. Autoradiographic observations in the dis- tribution and metabolism of N-[14C] nitrosonomicotine in mice. J. Cancer Res. Clin. Oncol. 98: 233-242, 1980. 77 (36) Waddell, W.J., and Marlowe, C. Localization of [14C] nitrosonomico tine in tissues of the mouse. Cancer Res. 40: 3518-3520, 1980. (37) Brittebo E.B., and Tjl%lve, H. Formation of tissue-bound N-nitrosonor- nicotine metabolites by target tissues of Sprague-Dawley and Fischer rats. Carcinogenesis 2: 959-963, 1981. (38J McCoy, G.D., Chen. C.B., and Hecht, S.S. Influence of modifiers of MFO activity on the in vitro metabolism of cyclic nitrossmines. In: M.J. Coon, A.H. Conney, R.W. Estabrook, H.V. GeIboin, J.R. Gillette, and P.J. O'Brien (eds.). Microsomes, Drug Oxidations, and Chemical Carcinogene sis, Vol. II. New York, Academic Press, 1980, pp. 11841192. (39) McCoy, G.D., Katayama, S., Young, R., Wyatt, M., and Hecht, S.S. In- fluence of chronic ethanol consumption on the metabolism and car- cinogenicity of tebaccerelated nitro samines. In: H. Bartsch, I.K. O'NeilI, M. Castegnaro, M. Okada, and L. Davis &is.). N-Nitroso Compounds: Occurrence and Biological Effects. IARC Sci. PubL 41: 309318,1982. (4OJ Waddell, W.J., and Marlowe, C. Inhibition by alcohols of the car&o genicity of radioactive nitrosonomicotine in sites of tumor formation. Science 221: 51-52, 1983. (41) Chung, F-L., Juchatz, A., Vitarius, J., and Hecht, S.S. Effects of dietary compounds on target tissue alpha-hydroxylation of N-nitrosopyrrohdine and N'-nitrosonornicotine. Cancer Res. 44: 2924-2928,1984. (42) Chung, F-L., Wang, M., and H&t, S.S. Effects of dietary indoles and isothiocyanates on N-nitrosodimethylamine and 4-(methylnitro- saminokld3-pyridyl)-l-butauone alpha-hydroxylation and DNA meth- ylation in rat liver. Carcinogenesis 6: 534543, 1985. (43) Swarm, P.F. Effect of ethanol on nitrosamine metabolism and distribu- tion. Implications for the role of nitrosamines in human cancer and for the influence of alcohol consumption on cancer incidence. IARC Sci. Publ. 5? 501-512, 1984. (44) Manson, D., Cox, P.J., and Jarman, M. Metabolism of N-nitrosomorpho line by the rat in viuo and by rat liver microsomes and its oxidation by the Fenton system. Chem. Biol. Interact. 20: 341-354,1978. (45) Hecht, S.S., and Young, R. Metabolic alpha-hydroxylation of N-nitro somorpholine and 3,3,5,5,-tetradueteroNnitrosomorphoI.ine in the F-344 rat. Cancer Res. 41: 5039-5043, 1981. (46) Hecht, S.S. N-Nitroso2hydroxymorpholine, a mutagenic metabolite of N-nitroscdiethanokunine. Carcinogenesis 5: 1745-1747,1984. (47) Lofberg, B., and Tjgve, H. Tissue specificity of N-nitrosmorpholine metabolism in SpragueDawley rats. Food Chem. Ibxicol. 23 647657, 1985. (48) Linjinsky, W., Taylor, H.W., and Keefer, L.K. Reduction of rat liver car- cinogenicity of 4-nitrosomorphohne by alphadeuterium substitution. J. Natl. Cancer Inst. 57: 1311-1313, 1976. (49) Chung, F-L., Palladino, G., and Hecht, S.S. Reactions of N-nitrosomor- pholine metabohtes with deoxyguanosine and DNA. Proc. Am. Assoc. Cancer Res. 26: 89, 1985. 78 (50) Chung, F-L., and Hecht, S.S. Formation of the cyclic l,NZ-glyoxal- deoxyguanosine adduct upon reaction of N-nitroso-2-hydroxymorphe line with deoxyguanosine. Carcinogenesis 6: 1671-1673,1985. (51) Bartsch, H., Camus, A., and Malaveille, C. Comparative mutagenicity of N-nitrosamines in a semi-solid and in a liquid incubation system in the presence of rat liver tissue fractions. Mutat. Res. 37: 149-162, 1976. EXPERIMENTAL STUDIES INVOLVING EXPOSING LABORATORY ANIMALS TO SMOKELESS TOBACCO OR ITS CONSTITUENTS This section reviews bioassays evaluating the carcinogenicity in ani- mals of smokeless tobacco and its constituents, particularly the tobaccospecific nitrosamines (TSNA) described in the section on the chemical constituents of smokeless tobacco. The bioassays involved multiple routes of administration of chewing tobacco, snuff, or extracts of these products and of several TSNA. Studies of chewing tobacco, snuff, and TSNA are summarized in tables 1 to 3 respectively, with comments on the individual investiga- tions provided below. Bioassays With Chewing Tobacco Oral Administration An alcohol extract of Indian chewing tobacco diluted 150 (group 1) or 1:25 (group 2) was gavagefed to male Swiss mice over 15 to 20 months. In another group of mice, a mixture of the tobacco extract with stan- dard laboratory diet was administered over 21 to 25 months (group 3). This treatment produced tumors in 8 of 15 mice at risk in group 1, in- cluding 5 mice with lung tumors and 2 with liver tumors; 4 of 10 mice at risk in group 2 developed lung and liver tumors. The feeding experiment (group 3) resulted in 8 of 10 mice with tumors, specifically 4 with tumors of the lung and 4 with liver tumors. Despite the high toxicity of the tobacco extracts and certain short-comings of the methodology, these assays indicate that the extract of chewing tobacco is carcinogenic in mice (1). Application to the Oral Mucosa and Cheek Pouch Three different extracts of an Indian chewing tobacco were applied daily for up to 18 months to the buccal mucosa of strain A and Swiss mice. No excess of tumors was observed (2). The oral mucosa of a group of weanling Wistar rats was painted twice weekly with a 2-percent alkaloid-free extract of an Indian chewing tobacco. No tumors were observed at the application site even though applications were con- tinued throughout the lifespan of the rats 13). 79 E TABLE l.-Bioassays for Carcinogenic Activity of Chewing `lbbaceo or Chewing `lbbacco Extracts* Duration of Fraction of Animals With `lbmors Route of Species, Exposure Application sex `l&t Material and Dose (Months) EXpOSed Controls Reference Oral- intubation Oral-feeding Skin- topical Oral- swabbing Oral- swabbing Oral-pouch implantation Oral-pouch Oral-pouch swabbing Oral-pouch swabbing Subcutaneous injection mice, M mice, M mice, M+F mice, M+F rats (NS)$ hamsters UW hamsters (NS) hamsters, M hamsters. F mice (NS) extract diluted I:25 4/l/2 diluted 1:50 15-20 0.2% extract in diet 21-25 DMSO extract (dose ?) 21-22 extracts applied daily, dose not given 2% alkaloid-free extract, dose not given- + time 2cm3 plug up to 18 Lifespan up to 30 DMSOextract three times weekly DMSOextract three times weekly, dose not given 2% tobacco extract in water, twice daily application 2% tobacco extract partially 18-24 5 6 IO-23 4110t lung adenocarcinoma 8115f and liver carcinoma 8llOt lung adenocarcinoma 0110 011 no excess tumors compared to controls o/10 0112 o/50 o/20 l/20 0110 0114 o/12 0112 3117 1117 squamouscell carcinoma or completely free of alkaloids, 25 solution once a month (site not specified) o/7 0111 0110 o Abbmviskm: DMSO. dimethyl sulfoxide t Animals at risk. $ INS) = not stated. TABLE 2.iBioassays for Carcinogenic Activity of Snuff or Snuff Extracts* Route of AoDlication Species, Sex `ht Material and Dose Duration of Fraction of Animals With `hmors EXpOEUre R&r- Amdications lMontbsl EXDO!%Xl Controls ence Oral-feedinz Hamsters. M S. 20% of diet Once dailv 24 0/1OOt Oil00 17 Lies-oaintinn Mice. M SE. dose not liven 3 times dailv 2 o/20 0120 18 Oral-swabbing Rats, M SE+H 0.5 ml daily 2 SE (approx. 30%) 0.5 ml daily up to 30 SE (approx. 30%) 0.5 ml daily up to 30 + (NNN+NNK) NNN+NNK 0.5 ml daily up to 30 Lip canal- Rats, F S 200 mg twice daily 9-22 instillation S 200 mg twice daily 18 H 200 mg 18 S+H twice daily 18 Lip canal- Rats. M S 50 mg daily up to 30 instillation s+se ES 50 mg daily 50 mn daily up to 30 UP to 30 0120 o/30 5/30 (3 papilloma in oral cavity. 2 lung adenoma) 13/30 (8 papilloma in oral cavity, 5 lung adenoma) 1142t (oral carcinoma) l/l0 (oral carcinoma) 017 217 (2 oral carcinoma) 0120 18 l/21 (lung adenoma) 20 l/21 (lung adenoma) 20 1121 (lung adenoma) 20 o/20 Oil0 0110 0110 21 22 22 22 3132 (papilloma and 1 Oil0 20 carcinoma in test canal, 1 oral papilloma) 1132 (oral papilloma) Oil0 20 2121 (oral papilloma) Oil0 20 Cheek pouch- Hamsters 10 ml paste once up to 30 0150 0150 instillation lNS)S ? 6 0125 2: H ? 6 0125 27 SfH ? 6 11125 (papilloma and carcinoma 27 of the oral cavity) Subcutaneous Rats, M+F SE 50 mg. 84 weekly 26 0182 0182 28 injection applications Rats (NS) TE 45 mg. 70 weekly 21 + 4 18175 1175 29 aoolications * Abbreviations: ES, extracted snuff; H. infected with herpes simplex virus; NNK. 4.(methylnitrosamino)-I-(-3.pyrldyl).I-butanone: NNN. N'-nitrosonornicotine. S. snuff. SE, snuff extract t NO tumors of the oral cavity, esophagus. nasopharynx and larynx; all other tumors nearly identical to those in control animals. a3 $ INS) = not stated. TABLE 3.-Csrcinogenicity of Tobacco-Specific Nitrosammes* Nitrw Species and samine Strains Route of Principal Application lhrget Organs Doee NNN NNK NAT NAB NNA A/J mouse F344 rat SpragueDawley rat Syrian golden hamster AIJ mouse ip. F344 rat S.C. Syrian golden hamster S.C. F344 rat F344 rat Syrian golden hamster A!J mouse i.p. S.C. p.0. p-0. S.C. S.C. p.0. S.C. i.p. lung nasal cavity esophagus esophagus nasal cavity nasal cavity trachea nasal cavity lung nasal cavity lung, liver trachea, lung, nasal cavity esophagus none none 0.12 mmollmouse 0.2-3.4 mmokat 1.0-3.6 mmokat 8.8 mmoUrat 0.9-2.1 mmol/harnster 0.12 mmol/mouse 0.1-2.8 mrnokat 0.9 mmolhamatm 0.005 mmol/hamst.er 0.2-2.8 mmoUrat 3-12 mmolkat 2 mmol/hamster 0.12 mmollmouse ' Hoffmann and Hecht (11) A group of 12 male Syrian golden hamsters received topical applica- tions on the buccaI mucosa of a dimethyl suboxide (DMSO) extract of an Indian chewing tobacco three times weekly for 21 weeks. None of the treated hamsters developed tumors in the oral mucosa; however, 8 of 12 treated animaIs had 1eukopIakia. These changes were not seen in the oral mucosa of the animals treated with DMSO alone (4). In another bioassay, 12 male hamsters received applications to the cheek pouch of a DMSC extract of Indian chewing tobacco three times weekly over their entire lifespan. Tumors were not observed in the treated group or the control group (5). When 1 mg of a paste made of a chewing tobacco extract was applied topicaky to the mucosa of the cheek pouches twice daily over a 6-month period, and animals were maintained without fur- ther treatment for another 6 months, the incidence of hyperplasia in the buccaI pouches was 17.6 percent, that of dysplasia was 29.4 percent, and that of squamous ceil papilloma or carcinoma was 17.6 percent in 17 hamsters. There were no tumors in the 20 control animaIs (6). Fifty hamsters received implantations of a 2 cm3 plug of chewing tobacco in their cheek pouches. The opening of the cheek pouch was ligated and the animals were observed for 18 months. After 13 months, 21 of 50 animals had survived. No tumors were recorded upon termina- tion of the assays (7). Although the studies cited above had some inherent weaknesses due to short application time or low dose, it appears, nevertheless, that both the oral mucosa of rats and the cheek pouches of Syrian golden 82 hamsters are relatively resistant to the carcinogenic activity of the extracts of chewing tobacco. Subcutaneous Application Seventeen C57 black mice were subcutaneously injected with 1 ml of a 2-percent solution of either partly or completely alkaloid-free extracts of an Indian chewing tobacco once a month for 1 to 24 months. One squamous carcinoma at an unspecified site developed in one mouse receiving the partly alkaloid-free extract (8). Skin Application A large number of studies have been published regarding the tumori- genicity on mouse skin of various extracts of chewing tobacco. Most of these bioassays failed to produce skin tumors. The negative results ap pear to be due primarily to the low dose applied or the short duration of the applications (9,101. The negative results indicate also that the con- centrations of TSNA and PAH in these extracts do not suffice to induce tumors upon topical application (11). However, the application of meth- anol or DMSC extracts of cigarette tobacco induced a low but signifi- cant number of benign tumors in the skin of CAFl and Swiss mice when these extracts were applied three times weekly for up to 24 months to the shaved backs of the mice (12,13). A number of studies have reported tumor-promoting activity of the extracts of chewing tobacco when these were applied to mouse epidermis previously treated with a tumor initiator @&?,1416). The bioassay data with chewing tobacco are sum- marized in table 1. Bioassays With Snuff Oral Administration For 2 years, 50 male BIO 15.16 and 50 male BIO 87.20 hamsters were each maintained on a standard diet containing 20 percent moist, fresh snuff. Controls consisted of 50 male BIO 15.16 hamsters and 50 male BIO 87.20 hamsters on a diet containing 20 percent cellulose (of caloric value similar to the snuff-containing diet). The spectrum of tumors ob served was nearly identical in both groups. Hamsters of both strains gavaged 60 times with 5 mg of the carcinogen 3-methylcholanthrene (MC) had a significantly increased incidence of both benign and malig- nant tumors of the forestomach and large intestine. Hamsters of the BIO 87.20 strain also had an increased incidence of stomach cancers while the BIO 15.16 strain developed tumors of the skin. lo assay the cocarcinogenic activity of snuff, 50 hamsters of each strain received the diet containing 20 percent snuff plus 50 times 0.5 mg of MC. Compared to the control group (diet containing 20 percent cellulose), the tumor yield was not increased in the two experimental groups indicating a lack 83 of carcinogenic activity as well as of cocarcinogenic activity of the snuff in this setting (17). Application to the Lip, Oral Mucosa, or Cheek Pouch The upper lips of 20 male BALB mice were painted 3 times a day for 5 days weekly over a 2-month period with a concentrated water extract of snuff (group 1). In another group of 20 male mice, the upper lips were in- oculated with herpes simplex virus type 1 (HSV-1) and were subse quently painted with a concentrated snuff extract for 2 months (group 2). A control group of 20 male mice received inoculation of the upper lips with HSV-1 and painting with water (group 3). Wo months' exposure to snuff extract (group 1) or HSV-1 inoculation (group 3) alone did not induce dysplasia in the epithelium of the labial mucosa, while HSV-1 in- oculation combined with painting of snuff extract produced epithelial dysplasia and other histomorphologic changes (18). In respect to this and other studies in which animals are infected with herpes virus in addition to treatment with snuff extracts, it should be noted that 20 to 40 percent of the U.S. population have periodic occur- rences of labial herpes (19). Male F344 rats were treated for up to 30 months by swabbing the oral cavity with either a concentrated water extract of snuff (group 1; 13.2 vg NNN and 2.8 ug NNK per milliliter snuff extract solution), snuff ex- tract enriched with the tobaccc~specific nitrosamines NNN and NNK (group 2; 148 ug NNN and 30 ug NNK per milliliter snuff extract solu- tion), NNN and NNK alone in concentrations corresponding to those applied in group 2 (group 3; 135 pg NNN and 27.6 ug NNK per milliliter test solution), or with water alone (group 4). Groups 1,2, and 3 consisted of 30 male rats each and group 4 (control) of 21 rats. The incidence of tumors in groups 1 and 2 was not significantly increased over that in the control group. In the group of 30 rats treated with NNN and NNK alone, 8 animals had oral tumors (6 papillomas in the cheek, 4 papillomas in the hard palate, and 1 papihoma of the tongue), and 4 animals had lung carcinoma. This study indicates that snuff contains czucinogenic N-nitrosamines; however, when they are being tested in an admixture with other components in the water extract of snuff, their carcinogenic activity may be suppressed (20). A group of 21 male and 21 female SpragueDawley rats were treated with snuff placed in a surgically created canal in the lower lip. Approxi- mately 0.2 g of a standard Swedish snuff (pH 8.3) was given twice daily 5 days per week for 9 to 22 months. The mean retention time of the snuff in the canal was 6 hours, and the estimated daily dose was 1 g of snuff/kg b.w. Using the same methodology, another group of 5 male and 5 female rats was treated with alkaline snuff in the surgically created canal (pH 9.3). One of the 42 rats treated with regular snuff developed a squamous carcinoma in the oral cavity after 8.5 months. The exposure to the regu- lar snuff resulted in mild to moderate hyperplasia of the epithelium, 84 hyperorthokeratosis, and acanthosis. Among rats exposed to snuff for 18 to 22 months, 16 of 42 showed vacuolated cells penetrating deeper into the epithelium with hyperplastic and atropic lesions. Rats exposed to alkaline snuff differed little from those in the group treated with regular snuff. Outside the area of treatment, squamous cell hyperplasia of the forestomach was found in rats exposed to snuff for 18 to 22 months (21). In another bioassay using the same methodology as described by Hirsch and Johansson (21), the surgically created canal in the lower lip of F344 rats was filled five times each week over 28 months with either U.S. snuff (average 0.2 g per application; n=30), snuff enriched with its own water extract (n=30), or the extracted residue of snuff (n=21). Ten rats with the surgically created lip canal, and otherwise untreated served as controls. The incidence of nonspontaneous tumors in each group was the following rats treated with snuff had one squamous carcinoma of the oral cavity, one squamous cell papilloma of the hard palate, and one meningioma; treatment with enriched snuff led to one squamous cell papilloma of the floor of the mouth and one nasal olfactory tumor; treat- ment with extracted snuff induced one squamous cell papilloma of the hard palate. There were no tumors in the control group (20). Four groups of female SpragueDawley rats with surgically created canals in the lower lip, received the following treatments beginning at 3 months of age: group 1 was infected with herpes simplex virus type 1 (HSV-1) by scarification and topical application followed 10 days later by administration of snuff into the canal morning and night on 5 days per week; group 2 was infected with virus and received no other treatment; group 3 was sham-infected with sterile saline followed by snuff treat- ment; and group 4, not given virus or snuff, served as controls, The HSV-1 infection was repeated once after a l-month interval, and snuff treatment was continued for 18 months after which time all animals were killed. Three animals in each of groups 1 and 2 died from encephalitis shortly after the second infection with HSV-1. Squamouscell carcinomas of the oral cavity developed in two of seven rats, and a retroperitoneal sarcoma was seen in one of seven rats exposed to HSV-1 plus snuff. In the group exposed to snuff alone, 1 of 10 animals developed a squamous carcinoma of the anus and 1 of 10 a retroperitoneal sarcoma (22). In several studies, various forms of snuff were installed in the cheek pouches of Syrian golden hamsters for up to 20 months. The application of snuff did not lead to the induction of tumors in the cheek pouches nor at any other site of the oral cavity in any of these studies even though malignant tumors were induced in the oral cavity with high doses of 7, 12dimethylbenz(a) anthracene and 3-methylcholanthrene (z23-26). In an assay for the joint action of HSV and snuff, the buccal pouches of 125 Syrian hamsters were inoculated with HSV-1, HSV-2, or culture medium. The control and HSV inoculations were done once a month for 6 consecutive months. Then 25 hamsters with HSV-inoculated pouches received installations of commercial snuff twice daily into both the right 85 and left pouches. One month after the last HSV inoculation and 6 months after continuous snuff application, the assay was terminated. The buccal pouches were removed for histopathologic examination. Neither the application of snuff to the cheek pouches nor HSV infection alone induced neoplastic changes in hamster buccal pouches. However, HSV infection in combination with snuff resulted in epitheliaI dysplasia and in squamous carcinoma of the buccal pouches in 11 out of 25 ham- sters (27). This investigation provides the strongest evidence to date that snuff may increase cancer risk in animals; however, full evaluation is pre eluded since the findings have been published only in abstract form. Subcutaueous Administration A Swedish snuff was extracted with 6Opercent alcohol and resulted in 18percent dry extract, which was injected subcutaneously into rats with 7Opercent ethanol and tri-n-caprylin (1:ll as vehicle. The rats received a total dose of 4.2 g of extract with 84 weekly doses of 50 mg of extract. No tumors were observed at the area of injection (28). This result is quite different from an earlier one by the same investigators in which an alcohol extract from cigarette tobacco (20percent yield) was injected into 75 rats with 70percent alcohol and glycerol as solvent (1:3). Per week, 45 mg extracts were injected until the total dose amounted to 3.2 g/rat. After 25 months, 18 of 75 rats had developed malignant tumors, primarily sarcomas at the injection site (29). The bioassay data with snuff are summarized in table 2. Bioassays With Constituents of Smokeless Tobacco At least three types of carcinogens occur in smokeless tobacco: poly- nuclear aromatic hydrocarbons (PAH), polonium-210 (21oPo), and N-nitrosamines. One of the PAH identified in smokeless tobacco, benzo(a)pyrene (up to 72 ppb), has long been recognized as an animal carcinogen (18,24,30). Levels of ZloPo in processed tobacco amount to 0.1-1.0 pCi per gram and to 0.18-1.22 pCi/g in commercial U.S. snuff products. Ionizing radiation can cause multiple types of cancer in ani- mals and humans raising the possibility that the alpha-radiation of 2loPo may contribute to the carcinogenic potential of smokeless tobacco and especially snuff f31,31). Three groups of N-nitrosamines have been identified in smokeless tobacco. All of the 4 volatile nitrosamines thus far identified are carcino- genic in animals (3). These are nitrosodimethylamine (0 to 215 ppb), nitrosopyrrolidine (0 to 291 ppb), nitrosopiperidine (0 to 107 ppb), and ni- trosomorpholine (0 to 690 ppb). Seven nonvolatile nitrosamines have also been identified in smokeless tobacco. Of these, only nitrosodiethano lamine (30 to 6,800 ppb) is a known carcinogen in mice, rats, and hamsters (33) Swabbing of the oral cavity of 20 male and 20 female hamsters with solutions of these agents three times weekly for 45 weeks 86 (20 mg per application) induced tumors of the nasal cavity in 17 animals, tumors of the trachea in 6, and a tumor of the larynx in 1 of the hamsters (34). The most abundant carcinogens in smokeless tobacco yet identified are the tobaccospecific nitrosamines (TSNA). These are formed during the processing of tobacco from its alkaloids. So far, seven TSNA have been identified in smokeless tobacco. Of these, N `-nitrosonornicotine (NNN; 470-135,000 ppb) and 4-(methylnitrosamino)-1-(3-pyridyl)-l- butanone (NNK; 30-13,600 ppb) are powerful carcinogens in mice, rats, and hamsters (table 1; 1,9). Table 3 summarizes results from bioassays administering TSNA to test animals. A variety of tumors were produced, particularly in the esophagus, nasal cavity, and lung. In a recently com- pleted investigation, daily swabbing for up to 30 months of the oral cavity of F344 rats with a saline solution containing 135 ppm NNN and 28 ppm NNK led to the development of benign oral tumors in 8 and lung carcinoma in 4 of 30 rats. Neither oral tumors nor tumors of the lung were observed in the negative control group (20). This study sug- gests that NNN and NNK may be tumor&&c at the site of exposure as well as systemically. Full evaluations of these results are precluded, however, since the original manuscript is now under journal review and not published. It is noteworthy that some of the bioassays indicated that relatively low doses of the TSNA could induce tumors. In hamsters, a total dose of only 0.2 mmol/kg of NNK induced a significant incidence of tumors (3s), whereas in F344 rats, 60 subcutaneous injections of a total dose of 20 mg (0.33 mmokkg) of NNK induced tumors of the liver in 10, tumors of the lung in 13, and tumors of the nasal cavity in 6 of 30 rats. Subcu- taneous applications to 27 rats of the same molar dose (0.33 mmokkg) of nitrosodimethylamine resulted in 6 animals with tumors of the liver and 1 rat with a tumor of the nasal cavity (36). For NNN, high tumor inci- dences were produced in F344 rats by a total dose of 1.0 mmovkg (37). Based on daily use for 30 years of 10 g of snuff containing 3.1 ppm of NNK, the estimated NNK exposure of a snuff dipper would be approxi- mately 0.02 mmollkg. Exposure to NNN from the same brand would be 0.4 mmol/kg (figure 3, chapter 2). Hence, the bioassays indicate that exposures in the dose range actually experienced by long-term snuff dippers induce tumors in animals. This is a distinctive and potentially important finding, since for most chemical carcinogens their carcino genicity was detected following exposure at doses much higher than usually received by humans. Of the other five TSNA, besides NNN and NNK, N `nitrosoanabasine (NAB; lo-6,700 ppb) and 4(methylnitrosamino)-1-(&pyridyl)-1-butanol (NNAL; 140-300 ppb) were moderately active carcinogens, and N-nitro soanatabine (NAT; 300-338,000 ppb) was inactive when tested at the low dose level of 9 n-mol./kg (9,38). 87 Recently, two additional TSNA have been identified in snuff: 4 (methyhritrosamino)-4-(3.pyridyl)-1-butanone (1,300-1,800 ppb) and 4-(methyhritrosamino)-(3-pyridyl)butenel (10 ppb; 6). These two nitro w-nines have not yet been tested for carcinogenicity. Mutagenicity Assays and Other Short-Term Tests Chewing Tobacco Nicotinu rusticu is a tobacco variety that is widely cultivated and used throughout India. Its ethanol extracts induced mutations in Sahwn~lla typhimurium TA98 and in V79 cells of Chinese hamsters. The addition of S9 liver homogenate from Aroclor-pretreated rats enhanced the mutagenic effect. No mutations were induced in TAlOO, TA1535, or TAX38 in the presence of the S9 homogenate. This ethanol extract of tobacco also induced micronuclei in bone marrow cells of Swiss mice (1,39,40). An ethyl acetate extract of Indian chewing tobacco induced sister chromatid exchange (SCE) in human lymphocytes and in a human lymphoblastoid cell line. In the latter system, S9 rat liver homogenate enhanced the effect. When the tobacco extract was tested in the absence of the S9 homogenate it did not induce ouabain-resistance in Chinese hamster V79 cells. The same extract, another ethyl acetate extract, and an ethanol extract of tobacco induced cell transformation in Syrian hamster embryo cells (41&f). The incidence of micronucleated oral mucosa cells in 27 Indians using khani chewing tobacco was 2.2 percent (0.8-4.9 percent). The incidence of micronuclei in exfoliated cells of nonchewers of similar ethnic back- grounds and dietary habits was 0.47 percent (0.0-0.9 percent) (43). Snuff The residue of organic solvent extracts from a U.S. commercial snuff was dissolved in DMSO and tested for the induction of SCE's in human peripheral lymphocytes. The organic snuff extract induced significant SCE's with a 0.05 percent concentration in lymphocytes of one of three donors, with a 0.15 percent concentration in lymphocytes in two of three donors, and with a 0.5 percent concentration in lymphocytes of all three donors (44). Tobacco-Specific N-Nitrosammes (`I'SNA) Of the seven TSNA so far identified in smokeless tobacco, only NNN and NNK were also tested for genotoxicity in short-term tests. In the presence of a liver microsomal preparation from Aroclor-induced rats, NNN and NNK caused dose-dependent mutations in SuZmoneUu typhimurium TAlOO and TA1535. Increased mutation frequencies were observed in the case of NNN at 2.5 umol and at 5.65 timoUplate and in the case of NNK at 1.4 wokplate (45-47). 88 NNN and NNK at 10-3 and 10-2 molar concentration each induced unscheduled DNA synthesis in freshly isolated hepatocytes from adult rats (48). Summary Chewing tobacco and extracts from various chewing tobaccos have been tested by oral administration in mice, topical application to the oral mucosa of mice, rats, and hamsters, and by subcutaneous adrnin- istration and shin application to mice. The investigations failed to demonstrate significantly increased tumor production. Short applica- tion times and low-dose exposures, however, limit the evaluation of the carcinogenicity of chewing tobacco or its extracts. Bioassays of snuff have likewise generally shown no excess cancer, although some experi- ments suggest that it may cause oral tumors in rats and hamsters that are infected with herpes simplex virus. Among the chemical com- ponents of snuff, the tobaccospecific nitrosamines NNN and NNK are powerful carcinogens. The doses of NNN and NNK that produce tumors in experimental animals are close to the doses estimated from lifetime exposure among human snuff dippers. References (1) Shah, A.S., Sarode, A.V., and Bhide, S.V. Experimental studies on mutagenic and carcinogenic effects of tobacco chewing. J. Cancer Res. Clin. OncoL 109: 203-207,1985. (2) Mody, J.K., and Ranadive, J.K. Biological study of tobacco in relation to oral cancer. Ind. J. Med. Sci. 13: 1023-1037, 1959. (3) Gothoskar, S.V., Sant, S.M., and Ranadive, K.J. Effect of tobacco and Iime on oral mucosa of rats fed on vitamin B deficient diet. Int. J. cancer 12 424-429,1975. (4) Suri, K., Goldman, H.M., and Wells, H. Carcinogenic effect of a dimethylsulphoxide extract of betel nut on the mucosa of the hamster buccaI pouch. Nature 2%? 383-384, 1971. (5) Ranadive, K.J., and Gothoskar, S.V. Betel quid chewing and oral cancer: Experimental studies. In: H.E. Nieburgs ted.). Prevention and Detection of Cancer, Part I, VoL 2. New York, Marcel Dekker, 1976, pp. 1745-1766. (6) Rao, A.R. Modifying influences of betel quid ingredients on B(a)P- induced carcinogenesis in the buccaI pouch of hamster. Int. J. Cancer 3.5: 581-586, 1984. (71 Peacock, E.E., Jr., and Brawley, B.W. An evaluation of snuff and tobacco in the production of mouth cancer. Plast. Reconstr. Surg. 23: 628-635,1959. (8) Ftanadive, K.J., Gothoskar, S.V., and Khanolka, V.R. Experimental studies on the etiology of cancer types specific to India. A. Oral cancer, B. Kangri cancer. Acta Unio. IntematL Contra Cancrum 19: 634639, 1963. 89 (9) International Agency for Research on Cancer. Tobacco habits other than smoking: Betelquid and areca-nut chewing and some related nitrosamines. IARC Monogr. EvaI. Carcinog. Risk Chem. Hum. 37: 291, 1985. (10) Wynder, E.L., and Hoffmann. D. Tobacco and Tobacco Smoke. Studies in Experimental Carcinogenesis. New York, Academic Press, 1967, pp. 198-202. (11) Hoffmann, D., Hecht, S.S., Omaf, R.M., and Wynder, E.L. Nitrosonor- nicotine: Presence in tobacco, formation and carcinogenicity. IARC Sci. Publ. 14: 307-320, 1976. (12) Wynder, E.L., and Hoffmann, D. A study of tobacco carcinogenesis, X. `Rimor promoting activity. Cancer 24: 289-301, 1969. (13) Wynder, E.L., and Wright, G.A. A study of tobacco carcinogenesis, I. The primary fractions. Cancer 10: 255-271, 1957. (14) Bock, F.G., Moore, G.E., and Crouch, S.K. Tumor promoting activity of extracts of unburned tobacco. Science 145: 831-833.1964. (15) Bock, F.G., Shamberger, R.J., and Meyer, H.K. `Armor promoting agents in unburned cigarette tobacco. Nature 208: 584-585, 1965. (16) Van Duuren. B.L., Sivak, A., SegaI, A., Orris, L., and Langseth, L. The tumor-promoting agents of tobacco leaf and tobacco smoke conden- sate. J. Natl. Cancer Inst. 37: 519-526, 1966. (17) Homburger, F., Hsueh, S.S., Russfield, A.B., Laird, C.W., and Van Dongen, C.G. Absence of carcinogenic effects of chronic feeding of snuff in inbred Syrian hamsters. `Ibxicol. Pharmacol. 35: 515-521, 1976. (18) Park, N.H., Herbosa, E.G., Ninkian, K., and Shklar, G. Combined effect of herpes simplex virus and tobacco on the histopathologic changes in lips of mice. Oral Surg. 59: 154-158.1985. (19) Barker, R., Burke, J., and Zieve, P. (eds.). Principles of Ambulatory Medicine. Baltimore, Wilhams and Wilkins, 1983, p. 1074. (20) Hecht, S.S., Rivenson, A., Braley, J., DiBeIlo, J., Adams, J.D., and Hoffmann, D. Induction of oral cavity tumors in F344 rats by tobacco specific nitrosamines and snuff. Submitted (1986). (21) Hirsch, J.M., and Johansson, S.L. Effect of long-term application of snuff on the oral mucosa: An experimental study in the rat. J. Oral Pathol. 12: 187-198, 1983. (22) Hirsch, J.M.. Johansson, S.L., and Vahlne, A. Effect of snuff and herpes simplex virus-l on rat oral mucosa: Possible associations with the development of squamous cell carcinoma. J. Oral Pathol. 13: 52-62, 1984. (23) Dunham, L.J., and Herrold, K.M. Failure to produce tumors in the cheek pouch by exposure to ingredients of betel quid; histopathological changes in the pouch and other organs by exposure to known carcino gens. J. Natl. Cancer Inst. 29: 1047-1067, 1962. (24) Dunham L.J., Muir, C.S., and Hamner, J.E., III. Epithehal atypia in hamster cheek pouches treated repeatedly with calcium hydroxide. Br. J. Cancer 20: 588-593, 1966. 90 (2.5) Dunham, L. J ., Snell, K.C., and Stewart, H.L. Argyrophihc carcinoids in two Syrian hamsters (Mesocricetis uumtus). J. Natl. Cancer Inst. 54: 507-513, 1975. (2s Homburger, F. Mechanical irritation, polycyclic hydrocarbons, and snuff. Effects on facial skin, cheek pouch, and oral mucosa in Syrian hamsters. Arch. PathoL 91: 411-417, 1971. (27) Park, N.H., Herbosa, E.G., and Sapp, J.P. Oral cancer induced in hamsters with herpes simplex infection combined with simulated snuff- dipping (Abstract 10). International Herpes Virus Workshop, Ann Ar- bor, August 11-16, 1985, p. 297. (28) Schm%hl. D. Priifung von Kautabakextract auf cancerogene Wirkung bei Batten. Anneimittel-Forsch. 15: 704-705, 1965. (29) Dmckrey, H., S&m&I, D., Beuthner, H., and Muth, F. Vergleichende Priifung von Tabakrauch-Kondensatn, Benzopyren und Tabakextract auf carcinogene Wirkung bei Batten. Naturwissenschaften 47: 605-606, 1960. (30) Campbell, J.M., and Lindsey, A.J. PolycycIic aromatic hydrocarbons in snuff. Chem. Ind. London 951, July 6,1957. (31) Harley, N.H., Cohen, B.S., and no, T.C. Polonium-210. A questionable risk factor in smoking-related carcinogenesis. Banbury Report 3: 93-104, 1980. (32) Hoffmann, D., Harley, N.H., Fisenne, I., Adams, J.D., and Brunnemann, K.D. Carcinogenic agents in snuff. J. Natl. Cancer Inst. (in press). (33) International Agency for Cancer Research. Monograph on the evahra- tion of the carcinogenic risk of chemicals to humans, VoL 17. Some N-nitroso compounds. Lyon, France, International Agency for Cancer Fitsearch. 1978, p. 365. (34) Hoffmarm, D., Rivenson, A., Adams, J.D.. Juchatz, A., Vinchkoski. N.. and Hecht, S.S. Effects of route of administration and dose on the car- cinogenicity of ZV-nitrosodiethanolamine in the Syrian golden hamster. Cancer Res. 4.X: 2521-2524, 1983. (35) Hecht, S.S., Adams, J.D., Numoto, S., and Hoffmann, D. Induction of respiratory tract tumors in Syrian golden hamsters by a single dose of 4-(methyInitrosamino)-l-(3-pyridylbl-butanone (NNK) and the effect of smoke inhalation. Carcinogenesis 4: 1287-1290,1983. (36) Hecht, S.S.. `B-u&in, N., Castonguay, A., and Rivenson. A. Compara- tive tumorigenicity and DNA methylation in F344 rata by 4-(methyi- nitrosamino)-l-(3-pyridyl)-1-butanone and N-nitrosodimethylamine. Cancer R.-es. 46: 498-502.1986. (37) Hoffmann, D., Rivenson, A., Amins, S., and Hecht, S.S. Dose-response study of the carcinogenicity of tobacco-specific N-nitrosamines in F344 rats. J. Cancer Res. CIin. Oncol. 108: 81-86, 1984. (38) Hoffmann, D., and Hecht, S.S. Perspectives in cancer research. Nicotine-derived N-nitrosamines and tobaccorelated cancer: Current status and future directions. Cancer Res. 4.5: 935-944,1985. 91 fW Bhide, S.V., Shah, A.S., Nair, J., and Nagarajrow, D. Epidemiological and experimental studies on tobacco related oral cancer in India. IARC Sci. Publ. 57: 851-857, 1984. f4OJ Sbimame, L.P., Monon, M.M., and Bhide, S.V. Mutagenicity of betel quid and its ingredients using " test systems. Carcinogens sis 5: 501-503, 1984. (41) Umezawa, K., Fujie, S., Sawamur, M., Matsushima, T., Koath, Y, Tanaka M., and Takayama, S. Morphological transformation, sister cbromatid exchange and mutagenicity assay of betel constituents. `Ibx- icol. Lett. 8: 17-22, 1981. (421 Umezawa, K., `Ibkayama, S., Fujie, S., Matsushima, T., and Sugimura, T. In vitro transformation of hamster embryo cells by betel tobacco ex- tracts. !IbxicoL Lett. 2: 243-246,1978. (4.3) Stich, H.F., Curtis, J.R., and Parida, B.B. Application of the micro- nucleus test to exfoliated cells of high cancer risk groups: Tobacco chewers. Int. J. Cancer 30: 553-559, 1982. (44) Tucker, J.D., and Ong, T. Induction of sister chromatic exchanges by coal dust and tobacco snuff extracts in human peripherai lymphocytes. Environ. Mutagen. 7: 313-324. 1985. 145) Andrews, A.W., Thibault, L.H., and Lijinsky, W. The relationship be- tween mutagenicity and carcinogenicity of some nitrosamines. Mutat. Res. 51: 319-326. 1978. (46) Bartsch, H., MaIaveiIIe, C., Camus, A.M., Mart.&PIanche, G., Brun, G., Hautefeuille, A., Sabadie, N., Berlin, A., Kuroki, T., Drevon, C., PiccoIi, C., and Montesano, R. Validation and comparative studies on 180 chemicals with S. typhimurium and V79 Chinese hamster cells in the presence of metabolizing systems. Mutat. Res. 76: l-50, 1980. (47) Hecht, S.S., Lin, D., and Castonguay, A. Effects of alphadeuterium substitution on the mutagenicity of 4-(metbylnitmsamino)-l-(3-pyridyl)- 1-butanone. Carcinogenesis 4: 305-310, 1983. (48) Williams, G.M., and Laspia, M.F. The detection of various nitre samines in the hepatocyte primary culture/DNA repair test. Cancer L&t. 6: 199206, 1979. CONCLUSIONS 1. The scientific evidence is strong that the use of smokeless toham can cause cancer in humans. The association between smokeless tobacco use and cancer is strongest for cancers of the oral cavity. 2. Oral cancer has heen shown to occur several times more fre quently among snuff dippers than among nontobacco users, and the excess risk of cancers of the cheek and gum may reach nearly fiftyfold among long-term snuff users. 3. Some investigations suggest that the use of chewing tobacco also may increase the risk of oral cancer. 92 4. Evidence for an association between smokeless tobacco use and cancers outside of the oral cavity in humans is sparse. Some investigations suggest that smokeless tobacco users may face in- creased risks of tumors of the upper aerodigestive tract, but results are currently inconclusive. 5. Experimental investigations have revealed potent carcinogens in snuff and chewing tobacco. These include nitrosamines, poly- cyclic aromatic hydrocarbons, and radiation-emitting polonium. The tobacco-specific nitrosamines N-nitrosonornicotine and 4-(methylnitro samino)-1-(3-pyridyl)-1-butanone have been de t.ect.ed in smokeless tobacco at levels 100 times higher than the regulated levels of other nitrosamines found in bacon, beer, and other foods. Animals exposed to these tobaccospecific nitro samines, at levels approximating those thought to be accumu- lated during a human lifetime by daily smokeless tobacco users, have developed an excess of a variety of tumors. The nitro- samines can be metabolized by target tissues to compounds that can modify cellular genetic material. 6. Bioassays exposing animals to smokeless tobacco, however, have generally shown little or no increased tumor production, although some bioassays suggest that snuff may cause oral tumors when tested in animals that are infected with herpes simplex virus. RESEARCHNEEDS It has been established beyond reasonable doubt that smokeless tobacco use can in- the risk of cancer. The experimental and epi- demiologic evidence is strongest for the association between oral cancer and the chronic use of snuff. Additional studies are needed to determine whether the patterns of risk differ according to the form of smokeless tobacco, including reseamh evaluating cancer risks that are associati with chewing tobacco and dry versus moist snuff, and to quantify fur- ther the levels of risk in relation to differing levels of smokeless tobacco expose. The influence of smoking, alcohol, and other factors (including viral expmues) on the smokeless tobaccoassociated risk of oral cancer also should be explored further with an emphasis on detecting possible inter- actions between these factors and smokeless tobacco. Inhaled snuff may increase the risk of nasal carcinoma. The feasibil- ity of initiating studies in areas where snuff sniffing is common should be ascertained, and studies should be launched to confirm and quanti- tate this possible relationship. There have been few studies of smokeless tobacco and esophageal, laryngeal, and gastric cancers. These investigations have provided equivocal results, but in the aggregate, their findings raise the possibil- 93 ity of some increase in risk among smokeless tobacco users. Additional case-control studies of these neoplasms should be encouraged. These studies should be large enough to assess the risks that are associated with smokeless tobacco use while controlling for the potential con- founding effects of smoking, alcohol, and other risk factors. Isolated reports have associated smokeless tobacco with cancers of the cervix, pancreas, and other anatomic sites. Investigators with exist- ing data from case-control studies of these neoplasms should be encour- aged to perform analyses to determine whether associations with smokeless tobacco exist. Similarly, existing data from cohort studies with information on smokeless tobacco use should be analyzed. Reports from two relatively large cohort studies have been published only as ab- stracts. These should be expanded with detailed descriptions of both the methods used and the findings for various cancers and should be up dated to include followup into the 1980's. Recommendations for addi- tional studies of the role, if any, of smokeless tobacco in the etiology of cancers outside of the upper aerodigestive tract should await the results of these analyses. On the basis of current knowledge, it can be assumed that chewing tobacco and snuff contain several unknown nitroso compounds that may be contributors to the carcinogenic potential of these products. In- depth analytical studies are needed for the identification of these unknown compounds. Furthermore, mechanisms of their in vitro and endogenous formation should be studied together with those of the ni- troso compounds that are already known to occur in smokeless tobac- cos. For the validation of the uptake of the major carcinogens by to bacco chewers and snuff dippers, markers should be measured in the target tissues and in physiological fluids. Major emphasis should be placed on the identification and assays of DNA-adducts with tobacco specific compounds in tissues of the oral cavity. Finally, trends over time in agespecific oral cancer incidence and mortality rates should be monitored to determine whether the increas- ing use of smokeless tobacco by Americans is influencing national or regional cancer patterns. Changes in the prevalence of use and in the characteristics of smokeless tobacco products should also be docu- mented. Such monitoring will provide a base upon which future investi- gations of associations between smokeless tobacco and cancer can be built. 94 Chapter 3. NONCANCEROUSAND PRECANCEROUS ORAL HEALTH EFFECTS ASSOCIATED WiTH SMOKELESS TOBACCO USE CONTENTS Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . ...99 The Effects of Smokeless Tobacco Use on Oral LeukopIakia/MucosaI Pathology and the %ansformation of Oral Soft Tissues ,107 Oral LeukopIakia/Mucosal Pathology . ,107 %ansformationofOraISoftTissues.. . . ,113 The Effects of Smokeless Ibbacco Use on the Gingiva, Periodontal Tissue, and salivary Glands ........ ,123 Background and Definitions ...................... ,123 Gingival and Periodontal Tissue ................... .123 SalivaryGlands.. ............................ ..12 6 The Effects of Smokeless `Ibbacco Use on lb&h .......... ,128 Background and Definitions ...................... ,128 DentaICaries ................................ ..12 9 OtherHardTissueEffects ....................... ,130 C0ncIusions . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . ...130 ResearchNeeds....................................131 References.............. . . . . . . . . . . . . . . . . . . . . . . . . ..132 97 This chapter addresses the health effects of smokeless tobacco use on the oral tissues through a systematic review of the relevant scientific literature of animal and human studies. The major areas addressed are the effects of smokeless tobacco use on the oral soft tissues, the periodontium, and the teeth. This chapter also reviews information regarding the potential of oral tissue altered by smokeless tobacco use to transform to dysplasia and malignancy. Within each area, except for the section on the transformation of oral soft tissues, those tissues or conditions that are suspected to be most af- fected by smokeless tobacco use, or that hold the greatest potential for health effects, are considered initially. Where contradictory evidence exists, these date are also presented. Studies that were judged to meet stringent selection criteria* are presented first, followed by data from less rigorous study designs and case reports. Within the section on the transformation of oral soft tissues, the pre sent&ion of the evidence is grouped according to clinical reports, cohort studies, and case-control studies. This was done so as to be consistent with the format used in the chapter on Csrcinogenesis Associated With Smokeless `Ibbacco Use (chapter 2). In some cases, studies referenced in this chapter are the same as those used in chapter 2. The reader should review both chapters to obtain all pertinent information contained in these studies. Only studies from the United States and Scandinavia are included for the sections on oral leukoplakia/mucosal pathology, gingival and perio dontal tissues, and salivary glands. This assures that studies dealing with similar types of smokeless tobacco are used for comparison pur- poses. However, the section on the transformation of oral soft tissues includes a fuller range of studies that have reviewed the histopathologic changes associated with smokeless tobacc&nduced lesions. Studies in- vestigating the histopathologic transformation of nonsmokeless tobaccuinduced lesions have not been included. A summary of selected studies that addresses study sample, methods, and observations is provided in table 1 as a ready alphabetical reference to the text. In addition, a summary of selected case reports is provided in table 2. Emphasis has been placed on the issues of preva- lence of oral tissue changes, types of changes, sitespecificity of changes, and the effects of doseresponse. o See Introduction. Overview. and Conclusions for discussion of criteria for causality 99 TABLE I.-Selected Study Summaries for the Noncancerous Oral Health Effects From the Use of Smokeless Tobacco Study Sample Methods Observations cmnmenta AxklL 1976 o 20,333 Swedes: o ????????????*?o 51% females, design. 49% males. o Data collected o Ages 15 years on tobacco and older. habits, medications taken. oral hygiene status, and prosthetic status. o ?o?*???o examinations utilized diagnosis based on specific clinical criteria. . Photographic documentation of all lesions diagnosed as leukoplakia or lichen planus. o Tissue speck- n-lens taken of selected ca3es. o Statistical analysis con- ducted: t-tests, chi square teats. and. if appropri- ate, Fisher's exact test. o Of 1,444 snuff users, 116 (8%) had "snuff dip per's lesion" (oral leuko plakia). o The prevalence of oral leukoplakia was 3.6% among the total population examimd o It is not clear how many of the snuff users were also tobacco smokers. o snuff dipper's lesion implies mumsal tissue changeaatthe site of snuff placement. Greer and Paulson. 1983 o 1.119 teenagers in grades 9-12. o 117 110.5%) smokeless tobacco users: 113 males. 4 females. o Denver. Colorado. o Cross-sectional design. o Questionnaire administered to determine years of use, frequency of use. brand of tobacco used. site of applica- tion, use of other confounding agents, and dental care history. LA?ukopl&i% Mucosal Pathology o A suggested association between level and duration of smokeless tobacco use and mucosal lesions (42.7% of smoke less tobacco users had oral mucosal lesions). o An analysis of the influence of cofactors was not conducted. o No statistical analyses reported. . Examiners blind to responses on questionnaire. o No comparisons reported between users of smokeless tobacco and nonusers. 100 TABLE I.-Continued Study Sample Methods Observations Comments Grew and Paulson. 1983 kont.) o ?o?*???o examination conducted of soft and hard oral tissues. o 26% of smoke less tobacco users had site specific gingival recession. o Lesions graded o Users with according to a lesions had scale developed longer use and by Ax&ll et al. higher daily (1976) and mcdi- exposure than fied by Greer users without and Pouleon. lesions. Teeth . `. found no evidence of tobacco associated dental ties." o No evidence of occlusal or in&al abrasion. o OnecaseOf cenical erosion. o Smokeless tobacco associated periodontal degeneration defined. o Did not assess the interrelation- ship of smoke less tobacco, cigarettes, and alcohol. Greer et al.. 1986 Salivary Glands o 45 smokeless o Cross-sectional o Of 18 tissue mbscco users design . Authors suggest samples with (43 males and 2 that the degree females); 15 sub- o Lesions graded salivary glands, of salivary gland jects in each by classification 4 demonstrated fibrosis. degan- developed by sialadeuitis and goup known as erative change, Grem and degenerative and sialadenitis juvenih, young Paulson. 1983. Changes. may be aas& adulta. and ated with geriat.liC. o ?????*?? ?*o? o A routine pattern of tobacco brand . Ages 13.74 lesions classified according to cllronic instead of a Y-. sialadenitis was generaJizd scheme. response awed o ??*???? not shown for Colorado. o Histomorpho any of the three by all tobacco. logical methods age groups. used on tissue Specimens. o Four patients (age3 21. 25. 50 * No statistical and 66) showed analysis either mild, conducted. moderate, or severs salivary gland fibrosis. 101 TABLE l.-Continued Study Sample Methods Observations Comments Hirsch et al.. 1982 LWk0phkid MUcosal Pathology o 50 male habitual o Cross-sectional snuff dippers. o Interpretation of o Dose considera- design. histomorphologi- tions were made o 41.3.year mean o Subjects Cal and histo chemical results and confounding age hmge 15-84 classified on a variables con- Years). four-degree scale demonstrated sidered. of lesion severity that the oral o ?????*? o Differences in (developed by mucosaf reaction AxdI et al., to snuff-induced brand of tobacco used were taken 1976); biopsies hyperplasia in were taken. the basal cell into account. o Histomorpho layers. logical and his. . Lethal damage tochemical was found in methods con- surface layers. ducted on sub o Duration of use jects' tissue and daily expo snecimens. sure to smoke o Tobacco and alcohol use histories ascertained from a questionnaire. less tobacco were shown to affect the severity of the leukoplakia. o Dysplasia could not be predicted by using sug gested CIinical degree of lesion classification. o Tissue speci. o Statistical mans from 74% analysis con- of patients ducted: oneway included salivary analysis of vari- glands. ante and multi- ple comparisons using the Scheffe method. Salivary Glands o The salivary glands and ex- cretory ducts showed degener- ative changes of a more severe nature than found in the sur- face epithelium. o 42% of salivary glands demon- strated siafaden- itis and degsner- ative changes. o Weak oxidative enzyme activi- ties noted in acinic cells in salivary glands with sialadenitis and degenera- tive changes. o Some signs of metabolic atypia noted. o Markedl degen- erative c k nges seen in salivary glands associ- ated with the more severely, clinicdy classi- fied lesions. o Degenerative changes not spe- cificdy defined by authors. o Authors state that variations in degenerative changes of salivary glands may lx bemuse of differences in brands of snuff and snuff- dipping habits, 102 TABLE l.-Continued Study Sample Methods Observations comments Jungell and Mahnstr6m. 1985 o 441 military recruits. o Ages 17-19 yf%X% o Finland. o 48 (11%) were snuff users. o l&9-year mean age (range 17-2 1 years). o Cross-sectional design o ???????**???? administered to ascertain to bacco product use and drinking habits and fre quency of dental care. o ?o?*???o ?????*?? tion conducted. o Biopsies taken of 21 snuff users with lesions. . Besting and stimulated @ar- affin served as the stimulator) salivary excre tions measured. o Statistical a&y- sis conducted t-test. o 10 nonusers of snuff also mea sured for sali- vary excretions. salivary Glands o Resting salivary flow of snuff users was signif- icantly higher than that of nonusers. o ?????o???? ??o????? ?o?? was higher, but not significantly, among snuff users than among controls. o There was no difference in buffering capacity be tween the two groups. o Authors inter- pret difference in resting salivary flowtobea reaction to the presence of the local irritant snuff. Modk et al.. GighI and 1980 Period0ntal o 232 school ClIiIM 119 males, 113 females. o 13.5 years mean age. o 11% of males were regdsr snuff users. o Sweden. o ????????????*?o design. o ?*????????? about tobacco product use his- tory and oral hygiene prac- tices. o ???*???????? dental indices used t.0 measure changes in oral hygiene and periodontal conditions. o Dental caries assessed clini- cally and radio graphically. o Statistical analyses con- ducted cross tabulations, mul- tiple regression, and student's t-test. o TheUseOfSmlff demonstrated a significant relation to gingi- vitis after con- trolling for plaque. o Effects of snuff on the gingival tissue included both location of the snuff and as a predictor of gingivitis in general. o Authors state snuff use may influence gingi val tissue directly result- ing in gingivitis. o Examiners blind to reqmse3 from iuterview. 103 TABLE l.-Continued Study Sample Methods Observations Comments Offenbacher and Weathers, 1985 o 565 m&s from 5 schools. o 13.8.year mean age (range lo-17 years). o 75 (13.3%) smokeless tobacco users. o ???????? L4?UkOjlhkid MUcosal Pathology o Cross-sectional design. o Frequency of occurrence of o Questio rmaire used to obtain history of tobac- co product "se, dental visits, and social history. o Intraoral exami- soft .g ele\ !Pri Eke vi ; tissue hology was aificantly rated in users gqJfgkF 11 lesions). nation conducted using some stan- o No attributable dardized indices. risk for mucosal o Statistical anafy pathology in smokeless ses included: chi tobacco users square, odds who were free of ~ZilkJJt&~, gingivitis. S o Control group Used. Giagival and PeriodOIltal o No relationship between smoke less tobacco use and the preva- lence of gingivitis. o Prevalence of gingival reces- sion signifi- cantly elevated in smokeless tobacco users. o A si fYnificant attrr "table risk exists for gingi- val recession m smokeless tobacco users. Teeth o Smokeless to bacco users with gingivitis had si,gnificantly greater caries arevalence corn pared with non- users without gingivitis. o Prevalence of caries was signif- icantly greater in users wth gingi- vitis who used both snuff and chewing tobacco cornoared with nonusers with gingivitis or those who were gingivitis free. o Soft tissue pzs not o Method of selecting schools for subject ascertainment not described. o ??*???*??*? variables considered. o Smokeless to baCCOUSeis viewed as a co factor with the presence of gin givitis in pro- moting gingivaf recesskon. o No dinhI defi. nitions provided for the assess- ment of gingiti- tis or gingival recession. 104 TABLE I.--continued Study Sample Methods Observations Comment43 Peacock et al., 1960 o 1,338 employees of local textile miu. o North Carolina. o Cross-sectional design o ?*????????? about tobacco product use and given an oral examination. Leukopw MUcosal Pathology o Highly signifi- cant relation- ships between chronic snuff and tobacco use and oral leukoplakia development found for all age groups and for both sexes. . Examiners blind to interview rmponma. o 90% of employees had either poorly fitting complete dentures or only few and rxrious teeth o Many employees have had the habit since they were 3 years old. Pot&on et al, 1984 o 445 subjects: 52% females, 47% males . 56 (12.6%) smokeless to hacco "Sal3 (all -W. o 16.7-year mean age Irange 14-19 Y-L . Rurd Colorado. o Cross-sectional d@P. o ???????**???? ????*??????? (same as one usedinGrmx and Paulson. 1983). o ?o?*???o ?????? nation conducted oforalhardand soft tissues. o ?????*? ???? by classification developed by GWSIld Poufson. 1963. o Of 56 smokeless t,htwco users, 35 (63%) had lesions of the hard or soft timues. o 33 (58.9%) smokeless tobac- co users had mu- cosal alterations. o Mucod lesions were found in area of quid placement. o Duration of use and length of daily exposure were factors in the development of lesions. o Multiple lesions in&same subject reported Giivd and Paiodontal o Of 56 smokeless tobacco users. 15 (27%) had site specific gingival recession: 2 users had periodontal lesions only; 13 had both mucosal lesions and periodontal destruction. . Examiners blind to responsea on q"estionnaire. o Definitions of CIiuical states provided. o ????????*??? *??o???? *?? ???????? o Ahistoryofcon- folmdmg vari- ables obtained. Effects of variables not addressed statisticauy o Periodontal degeneration defined. . Effects of con founding vari- ables not addressed statistically. 105 TABLE Z.-Summary of Selected Case Report&i Study NUlUk Product Duration country of users Age Ueed of use Ffndings Archard E&r 1972 Chl-kte", Amlstro"g, and McDaniel. 1979 Christen, McDaniel. and Doran, 1979 Frithiof et al.. 1983 Hoge and Kirkham. 1983 Pindborg and Poutson. 1962 Pindborg and Re".Wlp, 1963 Zitterbart. Marlin, and Christen, 1983 USA USA USA USA De-k De-k USA 3 1 14 21 1 7 12 1 31 42 60 S"Uff Snuff S"Uff 11 years 20 years 50 years 36 Snuff 13 years 18-22 Snuff, 6 months chewing to tobacco 9 years 31-79 Snuff 1060 years 21121 with snuff- induced lesions local- idtoareawhere snuff was held; 2/21 with observable gingival retraction. 20 S"Uff lY- Gingival recession and hyperkeratosis found where t.obam was habitually placed. Not reported S"Uff 20-30 years 4/7 bad whitish mucous membrane with a delicately folded appearance at site of snuff placement. 3483 Snuff 20-50 years 12112 with mucous membrane that was "whitish, sometimes yellowish-brown, dry appearance with a very delicately folded or finely grooved surface." 36 Chewing tobacco 24 years Gingival recession, "smokeless tobacco users lesion," and abraded occlusal sur- faces of posterior teeth found where tobacco was habitually placed. A homogeneous eosin- opbilic submucoszd deposit above the "linor salivary glands did not initiate a" in- flammatoryrespo"= nor support the possi- bility that the deposits were amyloid. Gingival rcsession, clinical leukoplakia, period0"td bone loss, and tooth abrasion found where tobacco was habitually placed. 8114 with clinically detectable gin&al recession; 9114 with clinical leukoplakia; 11114 with erythema- tou.9 soft tissue changes where te bacco or snuff was habitually held. 106 THE EFFECTS OF SMOKELESS TOBACCO USE ON ORAL LEUKOPLAKWMUCOSAL PATHOLOGY AND THE TRANSFORMATION OF ORAL SOFT TISSUES Oral LeukoplakiaMucosal Pathology Background and Deftitions Various oral soft tissue effects of smokeless tobacco use have been relxxted in the literature. These effects include oral leukoplakia/mucosal pathology. The actual terms used and the definitions employed to describe these conditions vary widely from study to study (table 3). The World Health Organization (WHO) defines oral leukoplakia as a white patch or plaque that cannot be characterized clinically or pathologically as any other disease (1). The mucosal pathology that is found in smoke less tobacco users also has been referred to as hyperkeratosis, an oral mucosal lesion that exhibits an abnormal whitish (keratinized) appear- ance clinically. The authors' terms are employed when a specific study's findings are described. However, in the discussion portion of the report, the general terms of oral leukoplakialmucosal pathology are used. The association between smokeless tobacco use and oral leukoplakia/ mucosal pathology has been moderately studied. The WHO has stated that tobaux is an etiologic agent for the formation of oral leukoplakia (1). This association was n&firmed at an International Seminar on Oral Leu- koplalda and Associated Lesions Related to Tobacco Habits (21. In a re view of the effects of tobacco habits other than smoking, the use of smoke less tobacco/snuff was associated with the presence of leukoplakia (3). Studies in the United States Six studies have addressed the prevalence of oral leukoplakia/muco sal pathology in smokeless tobacco/snuff users (49). In two of these studies, blindness of the examiners toward the tobacco habits of the subjects was maintained, and oral tissue findings in smokeless tobacco users and nonusers were compared (7,9). Three of these studies investi- gated adults (4@ and three investigated adolescents (7,9). In addition, several case reports have described oral leukoplakialmucosal pathology findings in smokeless tobacco users (1@13). Highlights of these studies and reports are summarized below. Offenbacher and Weathers investigated the oral tissue effects of smokeless tobacco use in adolescent males from the greater metropoli- tan area of Atlanta, Georgia (9). They used oral examinations and self- administered questio maims on tobacco use. Of the 565 males who were examined, 75 (13.3 percent) used smokeless tobacco. The difference in the prevalence of mucosal pathology in smokeless tobacco users (22.7 percent) was statistically significant compared with that of nonusers (4.7 percent); however, the authors did not provide specific diagnostic 107 TABLE 3.-Variations in Terms Used and Definitions Provided for LeukoplakiaMwoeal Pathology Associated With Smokeless Tobacco Use by Studies Cited Study Term(s) Used Definition(s) Provided Comment.9 Ax6U 1976 Christ.S?n. Armstrong, and McDaniel, 1979 christen, McDaniel, and Doran, 1979 Frithiof et al., 1983 Greer and Paulson. 1983 Hirsch, Heyden, and Thilander, 1982 Snuff- dipper's lesion. Clinical leukoplakia. Leukoplakia. Snuff- induced lesion. Oral mucosal lesions (alterations) associated with the use of smokeless tobacco. Snuff- induced lesions. A fourcategory classifi- cation scheme based on tissue color, wrinkhng. and thickening was used. "Implies only the clinical feature of a white patch or plaque on the oral mucosa which will not rub off and which cannot be characterized clinically or histologically as any other specific disease." "Implies only the clinical feature of a white plaque on themucosa.. .I' "Tissue changes in the oral mucosa" that are due to snuff use. These lesions were defined by a modification of a clinical grading method developed by Axdl et al. 1976. These lesions were defined by the grading method developed by Ax&l et al., 1976. The authors believe that this is a well-defined irritation that excludes it from the diagnosis of leukoplakia. The authors cite the WHO 1978 and Waldron and Shafer 1975 references (l&I The authors cite the Waldron and Shafer 1960 reference (48). The authors cite the WHO 1978 reference for the definition of let&o plakia and state that "since the snuff-induced lesion, with its typical clinical pattern and its specific etiology, obvi- ously constitutes a definite diagnostic entity, the term `leukoplakia' is avoided. I' In addition, lesions were classified by their texture, contour, and color. - 108 TABLE 3.--contiuued Study lkrm(s) Used Definition(s) Provided Comments Hoge and Kirkham, 1983 Moore, Bissinger, and ProehL 1952 Oral leukoplakia. Offenbacher Mucosal and pathology, Weathers, soft tissue 1985 pathology. Peacock, Greenberg, and Brawley, 1960 Pindborg and Poulson, 1962 Pindborg and Renstrup, 1963 Poulson, Lindenrnuth, and Greer, 1984 Zitterbart, Generalized Marlin, and smokeless Christen, tobacco 1983 users lesion. Hyper- keratotic- appearing tissue. Leukoplakia. Leukoplakia. Snuff- induced leukoplakia. Oral mucosal lesions (alterations) associated with the use of smokeless tdacco. No definition is provided, although the authors dis- cuss the "formation of a hyperkeratotic zone in the region of the `snuff pouch' where the tobacco is habitually held." No definition provided. No definitions provided. "A pearly white plaque on the mucous membrane which could not be scraped off with a tongue blade." No definition provided. No definition provided. The clinical appearance of these lesions was defined by a grading method developed by Greer and Poulson, 1983. No definition provided. The authors cite the Shafer, Hine, and Levy 1969 reference 149). The pathological findings identified by the investi- gators included morsica- tio, ulcer, keratosisileuko plakia, vesiculobullous, petechiae, abscess, erythema. mucocele, and pericoronitis. The investigators described the mucous membrane as having a slightly whitish, deli- cately folded appearance. The investigators de scribed the leukoplakias as "slightly whitish, some times yellowish-brown. dry appearance with a very delicately folded or finely grooved surface." Alterations in texture, color, and contour of the mucosal lesions also were identified. The lesion was described clinically as "peculiarly wrinkled and thickened." 109 criteria in this assessment. The range of mucosal pathologic findings in- cluded such conditions as morsicatio (cheek biter's lesion), ulcer, kera- tosisileukoplakia, vesiculobullous, petechiae, abscess, erythema, mucocele, and pericoronitis. Although 50 percent of the smokeless tobacco users with mucosal pathology had keratosisfleukoplakia com- pared with 3.8 percent of the nonusers with mucosal pathology, the authors did not identify the locations of the mucosal pathologies. Peacock, Greenberg, and Brawley reported a significant relationship between chronic tobacco use and the presence of oral leukoplakia* in a study of 1,388 textile mill workers in North Carolina (5). The 362 employees who reported using smokeless tobacco had a significantly higher prevalence of leukoplakia (34 percent) than did the 457 nonusers (7.4 percent). In addition, the authors noted a direct leukoplakia and age effect. In a study conducted in Denver, Colorado, Greer and Poulson exam- ined 1,119 teenagers in grades 9 to 12 to assess the relationship between oral tissue alterations and the use of smokeless tobacco (7). Smokeless tobacco was used by 117 (10.5 percent) of these teenagers. Of these, 42.7 percent had oral mucosal lesions? in the area of tobacco placement. Forty-six percent of the teenagers with mucosal lesions also had con- comitant periodontal tissue degeneration.$ Poulson, Lindenmuth, and Greer examined a sample of 445 teenagers in five rural Colorado towns to assess the relationship betwen oral tissue alterations and smokeless tobacco use (8). Smokeless tobacco was used by 56 (12.6 percent) of the teenagers. Of these, 58.9 percent had oral mucosal lesions in the area of habitual tobacco placement. Concom- itant periodontal degeneration was noted in 39.4 percent of those with oral mucosal lesions. Contrasting the results of rural versus urban adolescent smokeless tobacco users, Poulson, Lindenmuth, and Greer suggested that the duration of use may be critical in the development of "oral lesions" (8j.s Those adolescents with oral lesions used smokeless tobacco longer (an average of 3.3 years in the rural and urban groups) than those without lesions in both the rural and urban groups (2.3 years and 2.2 years, respectively). In addition, the authors noted similar effects of different levels of smokeless tobacco use in daily exposure. Users with oral le sions were exposed 205 minutes per day in the rural group and 177 minutes per day in the urban group compared with users with no oral le sions (110 minutes and 53 minutes, respectively). Also, more than twice * Leukoplakia was defined as a "pearly white plaque on the mwous membrane nhlch could not be scraped off wth a tongue blade. t The authors used a mod~ficatmn of the classificatmn method that u'as developed by .&&I et al. that Identifies the oral mucosal lesion> according to color. wrinkling. and thickening 1141. $ The authors define this degeneratmn as "site-specific gm@val recession wth apical migration of the gingwa to or beyond the cementwnamel ~unctmn. with OF wthout clmical evidence of inflammation." $ The term "oral lesmn~' used here includes penodonral tissue degeneratmn and oral mucosal lesions. 110 as many marked oral mucosal lesions were identified in the rural population as in the urban population. Smith et al. examin ed a population of 15,500 snuff users by cytologi- cal, histological, and visual means fs). Of these users, 1,751 (11.3 per- cent) demonstrated oral mucous membrane changes. Although no defi- nitions were provided, these changes were described as "cloudy or gray glistening" areas having "wrinkled appearance(s)" and presenting "white or red granular appearance(s)." The authors reported that when snuff was withdrawn, the tissue returned to normal appearance. Moore, Bissinger, and Proehl investigated the relationship between tobacco use and oral cancer in male patients age 50 years and older who attended the General lnmor Clinic in Minneapolis, Minnesota (4). The authors noted that a significant number of the patients who manifested oral leukoplakia (18 of 23-78.3 percent) used smokeless tobacco. A to bacco user in this study was defined as a person who used the tobacco product for 20 or more years. Apparently, some of these 23 patients were also pipe, cigar, or cigarette smokers, although the exact number was not specified. The authors indicated that the most severe patches of leukoplakia were seen in patients who chewed "strong" tobacco and over a longer duration (no quantification reported). In most instances in which patients had stopped using smokeless tobacco, leukoplakia disappeared. Several case reports (table 2) have described oral leukoplakiaimucosal pathology at the site of smokeless tobacco/snuff placement (1013). These cases represent males of various ages with differing years of smokeless tobacco/snuff use. Hoge and Kirkham reported that in one patient, withdrawal of snuff resulted in a reversal of the hyperkeratotic lesions (12). Studies in Scandinavia Studies of smokeless tobacco from Scandinavia have investigated the prevalence of oral 1eukopWmucosal pathology in users (X-19). Axell found 1,444 smokeless tobacco users (predominantly men) in the 20,333 Swedes who were examin ed for soft tissue lesions (la. Of these users, 116 (8 percent) had "snuff-dipper's lesion" (see table 3 for definitions). The prevalence of oral leukoplakia among the total study population was 3.6 percent. Hirsch, Heyden, and Thilander f18) graded oral mucosal lesions on an established four-point scale (14) and correlated these findings with the snuff habits in 50 Swedes ages 15 to 84 years who used snuff routinely. Younger patients were found to have lower degrees of pathologic changes, while a significant predominance of older patients was noted with higher degrees. The authors reported that patients with oral mucosal lesions of the highest degree had used snuff an average of 34.7 years compared with the 9.2- to 13.6-year average for patients with lower degrees of pathologic changes. They also noted that patients with high degrees of pathologic changes dipped twice as long per day (an 111 average of 10.1 and 10.6 hours per day) as patients withlower degrees of pathologic changes (5.2 and 6.5 hours per day, respectively). Although these patients reported multiple tobacco habits, the authors stated that no differences in clinical grading were found between patients who used snuff only and those who used snuff and other tobacco products. In addition, several case reports have described oral leukoplakial mucosal pathology (table 2). In Sweden, Frithiof et al. examined 21 male snuff users ages 31 to 79 years (19). All had snuff-induced lesions that were localized to the area in the oral cavity where the tobacco was held. Similarly, leukoplakia lesions were found at the site of snuff place ment in all 12 male users of snuff ages 39 to 83 years in a study in Den- mark (15). In this latter study, 3 weeks after one of the patients discon- tinued snuff use, the clinical appearance of the mucous membrane had returned to normal. In another report, four of seven Danish male users of snuff exhibited leukoplakia at the site of snuff placement (16). Discussion The studies from the United States and Scandinavia demonstrate that oral leukoplakialmucosal pathology is associated with smokeless tobacco/snuff use. In two studies, a higher prevalence of oral leuko plakia/mucosal pathology was found in users compared with nonusers of smokeless tobacco-22.7 percent compared with 4.7 percent (9) and 34.0 percent compared with 7.4 percent (5). In all of these studies, be tween 8 and 59 percent of smokeless tobacco/snuff users were found to have oral leukoplakialmucosal pathology. It appears that the oral leukoplakialmucosal pathology noted in smokeless tobacco/snuff users is found commonly at the habitual site of tobacco/snuff placement. Using a similar grading classification for snuff-induced lesions (7,14), all of the mucosal pathology that was noted in four studies was at the site of habitual tobacco placement (7,8,17,18). Similarly, the majority of the oral leukoplakia/mucosal pathology that was described in the case reports was found where the tobacco/snuff was usually placed. The duration of use (in years) and daily exposure (in hours or minutes) to smokeless tobacco appear to be critical in the development and sever- ity of oral leukoplakiafmucosal pathology. Three studies using similar approaches to the definition of oral 1eukoplakAmuco.A pathology and to the measurement of exposure noted this effect (7,418). Only two studies were designed to study the concomitant findings of oral leukoplakia/mucosal pathology and other tissue changes. The authors reported that 39.4 (8) and 46.0 (7) percent, respectively, of smokeless tobacco users with oral leukoplakia/mucosal pathology also had periodontal tissue degeneration (gingival recession). These oral soft tissue changes also were found at the site of habitual tobacco placement. In several studies where individuals had stopped smokeless tobacco use, the oral leukoplakia/mucosal pathology disappeared (4,6,12,15). 112 Background and Defiitions The previous section that discussed smokeless tobaccoinduced leu- koplakia noted that clinically observable changes in soft tissue mor- phology do occur as a result of smokeless tobacco use. Smokeless tobaccoassociated lesions that have been traditionally classified as leu- koplakias (white lesions) offer varying clinical degrees of differentiation and may persist or progress with continued smokeless tobacco use. Additionally, some leukoplakias have been observed to resolve clinically upon the cessation of smokeless tobacco use. This section of the report addresses the transformation of oral soft tissues. It discusses the poten- tial for smokeless tobaccoinduced lesions to regress, persist, or continue to progress to lesions with higher mahgnant potential or to malignancy. There are varying clinical and histologic definitions in the scientific literature related to tobaccoinduced changes (transformation) of oral soft tissues. The following definitions represent those most frequently encountered. It will be noted when significant variation of these defini- tions occurs in studies cited: o Oral leukoplakia-a white patch or plaque that cannot be charac- terized clinically or pathologically as any other disease (1). o Snuff dipper's leukoplakia-a leukoplakia associated with the use of smokeless tobacco. These are further characterized as to differ- ing morphologic forms. o Erythroplakia-a lesion present as a bright red patch or plaque that cannot be characterized clinically or pathologically as any other condition, such as carcinoma or infection. o FVecancerous condition-a generalized state that is associated with an increased risk of cancer based on epidemiologic or histo logic evidence. o Precancerous lesion-a morphologically altered tissue in which cancer is more likely to occur than in its apparently n0rma.l counterpart. o Acanthosis-an increased thickness of the spinous cell layer of the epithelium. o Hyperkeratosis-an increased thickness of the keratinized layer of the epithelium. o Hyperparakeratosis-an increased thickness of a normally para- keratotic layer of the epithelium, i.e., surface cells with retained nuclei. o Hyperorthokeratosis-an incrased thickness of a normally kera- totic layer of the epithelium, i.e., surface cells without retained nuclei. 113 o Chevron keratinization-a keratinization pattern typified by verti- cal streaks of parakeratinization that extend to the epitheliaI sur- face and create surface irregularities by extensions of the outer sur- face layer. o Dysplasia-abnormal tissue development characterized by vary- ing numbers and degrees of morphologic cell changes that reflect grades of severity. o Dysplastic changes include the following - Pleomorphism in the size and shape of cells and their nuclei. - Abnormal numbers of cells undergoing mitotic activity (discrep ancy in maturation). - Atypical mitotic cells. - Cytoplasmic atypicalities (altered nuclear to cytoplasmic ratio). - Hyperchromasia. - Irregular nuclear borders. - Basal cell hyperplasia. - Loss of polarity. o Carcinoma in situ-a significant number of dysplastic epithelial cdl changes that extend from the basal layer to the surface layer without violation of the basement membrane. o Verrucous carcinoma-a clinically verruciform cancer of epithelial tissue that tends to be slowly and locally invasive with a metasta- sis and mortality potential that is lower than classic squamous cell carcinomas. The cells are well differentiated. o Squamous cell carcinoma-a cancer of the stratified squamous epi- thelium that has varying clinical appearances, is invasive, extends beyond the basement membrane, and has a great potential for metastasis. Evidence of the relationship between smokeless tobacco use and the transformation of oral soft tissues is represented by the following 1. Clinical reports describing tobacco habits of persons with graded oral lesions. 2. Followup (cohort) studies of tissue changes, including trans- formation to malignancy, among patients with leukoplakia. 3. Casecontrol studies or case series of oral cancer describing con- comitant leukoplakia. A review of the evidence in each of these study areas follows: Clinical Reports of Oral Lesions in Association With Smokeless Tobacco Use Hirsch, Heyden, and Thilander (18) graded oral snuff-induced mucosal lesions in 50 patients on a four-point scale according to criteria developed by Ax&l (14): 114 o Degree 1: A superficial lesion with a color similar to the surround- ing mucosa, slight wrinkhng, and no obvious thickening. o Degree 2: A superficial whitish or yellowish lesion with wrinkhng and no obvious thickening. o Degree 3: A whitish-yellowish to brown lesion with wrinkhng, intervening furrows of normal mucosal color, and obvi- ous thickening. o Degree 4: A marked whiteyellowish to brown lesion with heavy wrinkhng, intervening deep and reddened furrows, and heavy thickening. Snuff habits and drinking habits of the patients were obtained from questionnaires. Patients in the degree 4 category had been snuff dippers significantly longer than the rest of the patients. Also, patients in de grees 3 and 4 dipped approximately twice as long per day as did pa- tients in degrees 1 and 2. The daily exposure to snuff was significantly longer in degree 4 (10.6 hours) than in degrees 1 (5.2 hours) and 2 (6.5 hours). When total exposure was compared between the four clinical groups taking into account hours of use per day as well as years of use, significant differences were found. In this study, no significant differences could be found with regard to clinical grading and histologicai appearances between patients with multiple habits (snuff, smoking, and drinking) and those who only used snuff. The four clinical degrees of lesions exhibited an agedependent ef- fect with younger patients usually found in clinical degrees 1,2, and 3 and a significant predo minance of older patients noted in degree 4. Degree 4 lesions included an increased number of mitotic figures, edema, and slight to moderate mflammation compared with the other three degrees. Eighteen percent of the patients exhibited slight epithe lial dysplasia, and lesions with slight epithelial dysplasia were found in all categories. Patients in the dysplastic group had been snuff dippers longer on average (23.9 years) as compared with those without dyspla- sia (19.5 years). No case of moderate or severe dysplasia was noted. (The authors referenced the WHO Collaborating Center for Oral Precancer- ous Lesions as the definition for dysplasia (l).) AxelI, M&n&ad, and Sundstrom obtained biopsies of the oral mucosal lesions of 114 male dippers ages 20 to 88 years from a sample of 1,200 Swedish snuff dippers (14). Clinically, lesions were graded (degrees 1 through 4) based on color and morphology. Lesions of higher clinical degrees were associated with greater daily exposure to snuff in terms of hours and grams of exposure. All but one of the biopsies showed increased epithelial thickness. The outer layers appeared vacua lated with occasional remnants of cell nuclei. Lesions in degrees 3 and 4 had more pronounced surface layers. Acanthosis was evident in all of the clinical groups. None of the biopsies showed changes that were interpreted as cellular atypia or epithelial dysplasia. The cessation of 115 snuff dipping for a few days was reported to result in clinical regression of the lesions with loss of the vacuolated layer. Greer et al. reviewed clinically and histologically examined smokeless tobacc&nduced leukoplakias from 45 patients ages 13 to 74 years @O), following criteria that were previously established by Greer and Poulson (7/ as adapted from Ax&lL The vast majority of the mucosal lesions were corrugated, white, and raised. No evaluations for an inter- relationship between smokeless tobacco use, smoking, and alcohol use and clinical or histologic tissue changea were attempted. Histologic examinations for specific changes were reported. Dark celI keratino cytes characterized by a strong affinity for basic dyes and by electron density of their cytoplasm and nucleus and suggested as dedifferenti- ated precursors of a neoplastic keratinocyte were found in 17 of 45 cases. However, their presence was unrelated to the clinical degree of the lesion. While they have also been observed in leukoplakias that are associated with smoking (or other causes), the control group of nontobaccoinduced hyperkeratoses demonstrated dark cell keratinocytes in only 3 of 45 rxses. Chevron keratinization of the epithelial layer representing altered cellular maturation was present in 42 of 45 smoke+ tobaccoinduced leukoplakias but in only 4 of 45 control leukoplakia cases. Koilocytotic changea appearing as vacuolated epithelial cells that may obscure the cytoplasm or appear with pyknotic nuclei, which are often associated with inclusion of viral particles in epithelial cells, were present in 27 of 45 smokeless tobaccoinduced leukoplakias. In the entire sample of 45 cases, only 1 case of dysplasia (described as occuing in a long-term smokeless tobacco user) was identified Three of the following characteristics had to be present for a lesion to be characterized as dysplastic: o Loss of celIular polarity. o Basal cell hyperplasia. o Altered nuclearlcytoplasmic ratios. o Anaplasia. o Dyskeratosis. o Atypical mitoses. Because the dysplasia case also involved the use of alcohol and smok- ing, it is not possible to attribute its appearance solely to smokeless tobacco use. In a study of 21 Finnish military recruits ages 17 to 21 years, mucosal lesions corresponded to the site of snuff placement and included the alveolar and labial mucosa to varying degrees (21). The duration and in- tensity of snuff use for this specific group could not be determined from the study. Epithelial hyperplasia and acanthosis were universally found under the light microscope. Hyperorthokeratinization was noted in 12 cases, hyperparakeratinization in 9 cases, and Chevron-type keratiniza- 116 tion in 1 case. One case of mild epithelial dysplasia was noted that in- cluded atypical and increased mitoses and loss of basal cell polarity. The authors concluded that this suggests a positive relation between snuff dipping and malignant changes. Van Wyk biopsied 25 snuff-induced lesions from Bantu smokeless tobacco users whose lesions had existed from a few weeks to 40 years (22). Comparison biopsies were also taken from healthy parts of the mucosa in the users, from healthy mucosa in nonusers, and from other white lesions and squamous carcinomas. From the biopsies obtained from snuff users, 18 cases of acanthosis, 23 cases of parakeratosis, 5 cases of keratosis, and 4 cases with numerous mitotic figures, plea morphism, hyperchromatism, and an irregular basal cell layer were noted. Additionally, 11 showed a disrupted appearance of the basement membrane. Those not associated with inflammation were considered possibly to be premalignant. Epithelium featuring these characteristics has been referred to by some as "disquiet epithelium." Contrarily, the author stated that "the impression is gained that no relationship exists between oral malignancy and the use of snuff." This was based on the widespread use of snuff but the occurrence of only one case of alveolar or sulcular cancer (not in a snuff user) in the hospital during this study. Several investigators have described connective tissue changes in snuff-induced lesions. A hyalinized, eosinophilic material that occurs well below the epithelium and around the minor salivary glands or in a plane that is generally parallel to the epithelial surface has been reported by Pindborg et al. (16), Archard et al. (23), Axell et al. (14), and Greer et al. (20). The exact nature of and underlying explanation for the finding are not clear. Additionally, the role of such a histologic finding in the development or progression of premalignant or malignant lesions has not been identified. Cohort Studies Several investigations have followed persons with oral lesions for subsequent health outcomes. Smith reported the lo-year followup results on a group of patients with smokeless tobacco-induced leuko plakias (24). In the original study, oral cytologies were performed on 1,751 patients presenting with leukoplakias out of 15,500 snuff users (6). Results of the oral cytology e xamination consistently indicated only benign hyperkeratoses. * Biopsies were made of 157 leukoplakic lesions. However, no objective criteria for lesions selected for biopsy were of- fered. None of the biopsies showed changes consistent with dyskera- tosis or malignancy. These patients were followed with repeat cytology smears for 5.5 years. No additional significant mucosal changes were * The use of oral c of a high rate of fa E tology for detecring dysplastic changes in leukoplakic lesmns is less than satisfactory because e negative findmgs. The hy rkeratinized nature of leuko lakic lesions renders them resistant to the oral cytology scraping technique. CeUu r missed m/. r changes in deeper layers o P the epithehum would thus likely be 117 reported. In a subsequent 4.5-year followup (10 years total followup), periodic biopsies were done on 128 of the 157 patients who had originally received biopsies (24). The authors reported no dyskeratosis or carci- nomas in the followup study. The method of followup was not specified. Significant numbers of patients were lost, and the clinicel and histologic diagnostic criteria were not fully described. A prospective study of oral cancer among persons with oral leuko plakia or other possible precancerous lesions was conducted in the Emakulum district, Kerala State, India, as part of a lo-year followup to a much larger study of 50,915 adults in 5 rural districts of India (26). Among those individuals who had been diagnosed as having a leuko plakia during the original survey, there was a malignant transforma- tion rate of 9.711,OOO per year for those who only chewed tobacco. For those who both smoked and chewed, the rate was 5/1,000 per year, while no malignancies were reported for individuals with or without tobacco habits who had not had a previous oral lesion The transformation rates among those with lesions were much higher than rates reported in the United States or European studies. While these results are not directly comparable to United States or European studies since the tobacco chewed in India is a variable mixture of betel leaf, areca nut, slake lime, and coarse tobacco, they suggest that the persons with leukoplakia are at increased risk of oral cancer. Specific clinical morphotypes of leukoplakia demonstrated varying potentials for malignant transfor- mation: homogeneous, 2.27 percent; speckled, 21.4 percent; and ulcer- ated, zero percent. In a small study of English coal miners, 8 of 22 patients with leuko plakia who chewed tobacco were followed for 5 years (27). Five of the eight cases showed no advance in the lesions, and two showed regres- sion. The author does not specify whether these were clinical or histo logic determinations or whether the smokeless tobacco habit persisted in all cases. One lesion that had been regarded as benign showed some hyperorthokeratosis and acanthosis of the epithelium but with no more than "minor epithelial atypia." The clinical appearance of this lesion was reported to have regressed initially over an intermediate 2-year period despite continuance of the habit of tobacco chewing and smok- ing. Subsequent followup over a 2-year period indicated that the lesion had progressed to an exophytic squamous cell carcinoma. The site of the lesion was where the patient had held tobacco for 30 years. While the malignant transformation rate in the group of chewing tobacco- associated leukoplakias was 12.5 percent, the small numbers and high dropout rate limit the significance of the finding. Of significance was the unpredictable course of the malignant lesion, initially regressing and then transforming into a squamous cell carcinoma. In a Danish study, 32 patients with snuff-induced leukoplakias from a group of 450 patients with leukoplakia were observed for a median time of 4.1 years (28). Each patient had also used alcohol, with 17 per- 118 cent claiming daily use. Thirty-three biopsies demonstrated hyperplas- tic epithelium with hyperparakeratosis in 87 percent of the cases; haIf showed vacuolated cells. One initial case of epithelial dysplasia was found, and one carcinoma was found to develop from a nondyskeratotic leukoplakia over the followup period. This represents a rate of premalig- nant or malignant transformation of 6.2 percent for either dysplasia or carcinoma. In comparing the rate of development of dysplasia and car- cinoma from snuff-induced leukoplakias to nonsnuff-induced leuko plakias, the authors found no statistically significant differences. How- ever, the rate of transformation in both groups was higher than would be expected in individuals without leukoplakic mucosa. In an earlier report on a small sample of 12 white male snuff-using leukoplakia patients (use from 20 to 50 years), Pindborg and Renstrup did not find any malignant transformation (15). Biopsies were taken from sites where the snuff was held. All 12 showed unkeratinized hyper- plasia of the epithelium with a few deep streaks of parakeratosis and downgrowth and broadening of the rete pegs with the outer layers of cells being vacuolated and large. The authors state that snuff-induced leukoplakias are easily reversible. Based on the limited size of this sam- ple, definitive conclusions could not be made. Oral Lesions Concomitant With Oral Cancer Thme hundred and thirty-three patients with cancers of the buccal cav- ity and pharynx from the Robert Winship Memorial Clinic in Atlanta, Georgia, were compared with three control groups: a group with dis- eases of the mouth other than cancer or with no diseases; a group with cancer of sites other than the mouth, pharynx, or larynx; and a group without cancer and whose mouths were not examined-see chapter 2 (29). The authors, citing leukoplakia as a precancerous condition, found leukoplakias "more commonly in women with low grade squamous car- cinomas arising in the mouth and with multiple cancers. Snuff dipping was frequently associated with leukoplakia and low grade cancer aris- ing in the mouth." In a case-control study in Minnesota of cancers of the alveolar ridge, floor of the mouth, and buccal mucosa, it was noted that leukoplakias and cancers of the mouth were related to the use of snuff or chewing to bacco (4). The most severe leukoplakias were reported among those who used "strong snuff" (no definition was provided) and held the quid at the same site for many years. Patients who quit using smokeless to bacco reportedly had leukoplakias disappear in most instances. A number of patients had multiple primary carcinomas that were also specific to the site of quid placement. Cancer lesions were described as having developed slowly over a period of several years, although no evidence of periodic clinical or histologic assessment was provided. McGuirt reported on 76 oral cancer patients, most with carcinomas of the alveolar ridge or buccal mucosa, identified from the tumor registry 119 at the North Carolina Baptist Hospital who had a documented history of heavy smokeless tobacco use (30). Fifty-seven of these patients used snuff and reported no cigarette, pipe smoking, or alcohol habits. The range of use was from 10 to 75 years. Leukoplakias had previously been excised in 13.9 percent of the cases, and 47 percent had associated leukoplakias at the time of surgery. The author cited "panmucosal in- sult" from smokeless tobacco use as the cause of multiple lesions and recurrences-a type of field cancerization. From histologic evaluations of oral tissue among 23 Swedish patients with anterior oral vestibular cancer who were snuff users, leukoplakic lesions were noted outside the snuff-associated tumor in 5 (31). Lake plakia and multiple carcinomas occurred together with the snuff- associated lesion in three cases. Eleven of nineteen cases assessed for presence of candida were positive. The temporal relationship between candida and carcinoma was not ascertainable, nor was the potential etiologic role of candida. Rosenfeld and Callaway examined data from records at Vanderbilt University Hospital, Nashville General Hospital, and the office of Rosenfeld for cases of squamous cell carcinoma arising in the mucous membrane of the anterior twothirds of the tongue, the floor of the mouth, the gingiva, and the buccal area (32). A total of 525 cases were examined in users and nonusers of smokeless tobacco-300 occurred on the gingiva and buccal areas. Among women with cancer of the buccal or gingival area, 90 percent had a history of snuff use. While no periodic quantitative or qualitative assessment of the natural history of the cancers is provided, the authors do offer the following clinical impres- sion of snuff-induced lesions in their study: These carcinomas arising in the inner cheek and gingiva frequently start as leukoplakia. Progressive thickening, cornification, and even- tual cauliflower-like ulcerations ensue. All stages in the progressive disease may be seen in microscopic sections from a mere slight in- crease in the keratin layer, through carcinoma in situ to invasive malignancy. Twenty-five cases of histologically confirmed buccal gingival cancer in female snuff users were identified at the University of Arkansas Medical Center from 1950 to 1959 (33). Eleven cases occurred at buccal sites, 10 gingival, and 4 buccal and gingival. The patients (ages 44 to 84 years-mean 67.5) had a smokeless tobacco habit between 20 and 50 years. The lesions corresponded to the site of habitual tobacco placement. Leukoplakia was a concomitant lesion and had been pres- ent for many years. Bepeat biopsies of lesions were made over long periods in some of the patients. Leukoplakic lesions from other parts of the mouth often showed atypia. An evolution from leukoplakia to pseudoepitheliomatous hyperplasia to early squamous cell carcinoma was found. 120 Di6cu6sion In characterizing the role of smokeless tobacco use in the clinical and histologic course of oral lesions, there are several problems. First, oral leukoplakia should be considered a dynamic changing lesion of the oral mucosa (34). Lesions retain the potential to resolve, remain static, or progress depending on a variety of factors that may be either exoge nous (e.g., smokeless tobacco use) or endogenous (e.g., natural tissue defenses and repair potential). To achieve comparability of results among investigators, a standard system for gauging epithelial dysplasia is needed. Patients then could be followed prospectively to quantify the incidence of dysplastic change, incidence of transforma- tion from a dysplastic state to a cancerous state, or in some cases transformation from an apparently benign to a cancerous state. But ethical considerations do not ahow lesions to be monitored continuously from benign states to moderate and severe dysplasias and carcinoma in situ. The next best alternative would be to provide estimates of risk for malignant transformation based on empirical and clinical observations or at least to quantify descriptively the association that smokeless tobacco-induced lesions have with other lesions or other potential etiologic factors. The body of literature on smokeless tobaccoinduced lesions and their potential for malignant transformation allows for the development of a conceptual model of the natural history of smokeless tobacco-induced lesions (figure 1). This model is a composite of various prospective, retrospective, cross-sectional, and case studies that relate to smokeless tobacco-induced lesions, It depicts progressive changes that may occur in some individuals who are habitual users of smokeless tobacco and potential outcomes that could include death or disfigure- ment for some individuals who use smokeless tobacco for several dec- ades. The data are clear that habitual smokeless tobacco use can pro duce mucosaI lesions (see leukoplakia discussion). It is also clear that where groups of patients with smokeless tobaccoinduced leukoplakias have been followed for several years, cases of cancer have been identi- fied. Finally, when considering studies of oral cancers in habitual smokeless tobacco users, there appears to be a consistent finding of leukoplakias either having been previously excised in the area of habit- ual tobacco placement or being found concurrently with and in proxim- ity to oral cancers. In comparing studies on the transformation potential of smokeless tobacco-induced leukoplakias, it is found that different criteria have been used by various investigators in defining dysplastic changes. The number and nature of criteria that are considered and that are consid- ered adequate to classify a case as dysplastic are not consistent. Addi- tionally, the degree of agreement on diagnosis based on histology and clinical history between individuals has been shown to be quite variable. Pindborg, FLeibel, and Holmstrup tested the degree to which a group of 121 FIGURE 1.-A Conceptual Natural History of Oral Mucosal Changes Associated With the Use of Smokeless `Ibbacc~ Diagnostic Oral Tissue Level status Smokeless Tobacco Exposure Time A HEALTH CLINICAL Leukcplaklas Erythroplakias Smokeless Tobacco Hablt P-ooablllty Low Probabmlit\/ High D~sp~asx Changes I t t CLINICAL and De&h Powble Deatr? Highly HISTOLOGIC 01 Loss of Tissue Powble or Potent'al and Function o for Disfigurement * I Months to "ear 10+ Years 122 oral pathologists could agree on diagnoses where nine cases of epithelial dysplasia, carcinoma in situ, or initial squamous cell carcinoma were examined (35). Color photomicrographs and information on the topog- raphy of the biopsy were presented. The authors' diagnoses were based on the criteria that are described in the report from the WHO Interna- tional Collaborating Center for Oral Precancerous Lesions (1). The degree of agreement with the authors' diagnoses for the nine cases ranged between 10 and 78 percent. This could partially explain the range in prevalence and incidence of malignant transformation that is reported by various investigators. Other contributing factors in comparing studies could include differ- ent population groups in terms of age and gender and other confound- ing variables (e.g., smoking, alcohol use, and type of smokeless tobacco product used). Each of these limitations is suggestive of the type of research that is needed. THE EFFECTS OF SMOKELESS TOBACCO USE ON THE GINGIVA, PERIODONTAL TISSUE, AND SALIVARY GLANDS Background and Definitions Reports of gingivitis, gingival recession, and degenerative salivary gland changes associated with smokeless tobacco use are contained in the literature. As with the previous section on oral leukoplakia, the terms used and the definitions employed to describe gingivitis and gingival recession vary widely from study to study. `lhble 4 displays the variations found in the literature. As each study is described in the fol- lowing narrative, the authors' terms are employed. However, in the discussion portion of this report, the general terms of gingivitis and gin- gival recession are used. General definitions for these terms and for sialadenitis follow: o Gingivitis-This condition refers to clinically detectable acute or chronic mflammation, either local or general, of the gingiva. o Gingival recession-In general, this condition describes the apical migration of the gingiva with or without clinical evidence of inflammation. o Sialadenitis-Inflammation of the salivary glands. Gingival and Periodontal Tissue Studies that assess the relationship between smokeless tobacco use and gingival and periodontal tissue effects are limited. The literature consists of several cross-sectional studies in teenagers and a few case reports. 123 TABLE 4.-Variations in `krms Used and Definitions Provided for Gingivitis and Gingival Recession by Studies Cited Study Term(s) used Deftition(s) Provided Conunents Christen, Armstrong, and - McDaniel. 1979 Christen, McDaniel, and Doran. 1979 :OZYd 1983 ' Ho and I(lr ham, .f 1983 Mod&r. Lavstedt, and Ahlund, 1980 C$enbacher Weathers, 1985 Paulson. Lindenmuth, and Greer, 1984 Zitterbart, Marlin, and Christen, 1983 Gingival recession, periodontal gxket. and loss of veolar bone. Clinically detectable gingival recession. Tobaccoassociated periodontal degeneration and periodontal lesions. Gingivai recession. Gingivitisigingival inflammation. Gingivitis. Gingival recession. lbbacco-associated "Defined as site periodontal degener- ation (other terms specific gingival include periodontal recession with apical migration of the deterioration," and "localized periodon- gingiva to or beyond the cementoenamel tal degeneration junction, with or associated with the without clinical site of tobacco evidence of placement"). inflammation." Gingivitis, Gmgival recession. No definition provided. No definition provided. No definitions provided. No definitions provided. Defined as site specific gmgival recession wrth apical migration of the gingiva to or beyond the cementoenamel junction, with or without clinical evidence of inflammation." No definition provided. No definition provided. No definition provided. The tissue changes were described in zrofs by the - The authors defined the recession as having "exposed approxi- mately 5 mm of labial root surface" and having destroyed the "entire functioning border of keratinized gingiva." - - The gingival recession was "considered slight to moderate, ranging in l-4 mm apical migra- tion when present." - - The clinical findings were described for each tooth site involved. 124 Studies in the United States Three cross-sectional studies have investigated the relationship of gingival and periodontal tissue changes and smokeless tobacco use in teenagers in the United States (7-9). Offenbacher and Weathers exam- ined the effects of smokeless tobacco use on mucosal pathology, on the presence of gingivitis and gingival recession, and on dental caries status (discussed in next section) (9). Of the 75 smokeless tobacco users, the authors noted 72 percent with gingivitis and 60 percent with gingival recession. In those with gingival recession, 6.6 percent presented with recession in direct juxtaposition to the location of the tobacco place ment. The authors did not describe how many users of smokeless tobac- co had demonstrated combinations of these oral conditions. Also, no specific clinical definitions were given for the assessment of gingivitis or gingival recession, although the latter findings were described as "slight to moderate, ranging from 1 to 4 mm apical migration of gingi- val tissue." The higher prevalence of gingival recession among smoke less tobacco users (60 percent) as compared with that found in nonusers (14.1 percent) was found to be statistically significant. There were no statistically significant differences in gingivitis prevalence between smokeless tobacco users (72 percent) and nonusers (77.1 percent). Of 117 adolescent smokeless tobacco users in Denver, Colorado, Greer and Poulson noted that 25.6 percent had tobaccoassociated periodontal degeneration (7). As noted earlier, this condition was de fined as "sitespecific gingival recession with apical migration of the gingiva to or beyond the cementoenamel junction, with or without clini- cal evidence of inflammation." Concomitant mucosal lesions were noted in 76.6 percent of those who had periodontal degeneration (gingival recession). In a study of rural Colorado teenagers, Poulson, Lindenmuth, and Greer (8) described 26.8 percent of 56 smokeless tobacco users with peri- odontal degeneration (gingival recession) as defined by Greer and Poulson (7). Eighty-seven percent of these had concomitant mucosal lesions. Several case reports (table 2) describe the occurrence of gingival reces- sion and periodontal tissue destruction in individual smokeless tobacco/ snuff users (1@13). The patients in these case reports were males who ranged in age from 18 to 36 years with varying duration of the smoke less tobacco/snuff habit ranging from 1 to 24 years. Although not uni- versally found, gingival recession was usually noted, and the majority of patients presented with recession that was specific to the site where the tobacco/snuff was habitually placed. Periodontal bone loss at the site of snuff placement was described in another patient who used snuff for 13 years (10). In one patient, 3 weeks after cessation of snuff use, there was no regeneration of the lost gingi- val tissue, although, as noted earlier, the hyperkeratotic areas had dis- appeared (121 125 Studies in Sweden Mod&r, Lavstedt, and Ahlund studied the oral health effects of smoking and snuff use in 232 Swedish school children ages 13 to 14 years (119 boys and 113 girls) (36). Thirteen (11 percent) of the boys used snuff. The children were interviewed regarding their tobacco and tooth- brushing habits, and examin ers (blind to the interview results) clinically assessed the degree of gingival mflammation, oral hygiene, and the presence of calculus (discussed in the next section). Standardized in- dices were used to assess all oral conditions. Controlling for the presence of dental plaque, gingival inflamma tion was the only variable that was significantly different between snuff users and nonusers. Snuff use was directly correlated with the degree of gingival mflamma- tion. The gingival inflammation noted was related to the site of smoke less tobacco placement. Discussion The relationship of smokeless tobacco use and the health of gingival and periodontal tissue has received minimal study. Because of the variation in study designs and diagnostic criteria, comparisons between available studies are inappropriate. Thus the effects of smokeless tobac- co use on these tissues are not clearly understood. With regard to gingivitis, one cross-sectional study noted no differ- ence between users and nonusers (9). Another study, however, empha- sized that there was a significant difference between users and nonusers and that snuff use was directly correlated with the degree of gingival inflammation (36). Gingival recession is a common finding among users of smokeless tobacco/snuff. In the U.S. cross-sectional studies, gingival recession was found in 25.6 to 60 percent of teenage users (7-9). In the two Col- orado studies, all the gingival recession was specific to the site of to bacco placement (25.6 and 26.8 percent) (8). In the Georgia study, only 6.6 percent of the gingival recession was in the area of tobacco place ment (9). In addition, several case reports have identified gingival reces- sion at the site of habitual tobacco placement (1@13). Between 76.6 and 86.6 percent of smokeless tobacco users who had gingival recession also had concomitant mucosal pathology (7,s). These soft tissue changes were found at the site of habitual tobacco placement. Salivary Glands Smokeless tobacco or its components may contribute to degenerative changes and severe damage, such as undifferentiated carcinoma, to the salivary glands and excretory ducts of humans and mice (1&20,2437). In a study that assessed the formation of tobacco-specific nitrosamines from the major tobacco alkaloid nicotine, Hecht et al., reporting from the histologic evaluation, noted two undifferentiated carcinomas of the 126 salivary glands in two groups of mice that were given injections of nitrosonomicotine (NNN) in saline or trioctanoin (37). Because of the uncommonness of salivary tumors in strain A mice, Hecht et al. con- cluded that the tumors were probably a result of systemic administra- tion of NNN. SiaIadenitis and degenerative changes in minor salivary glands were found in 16 of 50 habitual snuff dippers with a greater number belong- ing to the groups that were classified clinically as having the most severe snuff-induced lesions (18) (table 1). The findings from this study included a decrease in oxidative enzyme activities and indications of metabolic aty-pia that were based on enzyme histochemical tests. The salivary glands appeared to manifest more damage than the oral epi- thelium from snuff use. Variations in degrees of effect may be attrib uted to the variations in snuff dipping habits and brands of snuff. In a recent study by Greer and his colleagues (20) (table l), 45 smoke less tobacco users ages 13 to 74 years were clinically and histomorpho logically assessed for the effects of smokeless tobacco on the oral tissues. Of 45 tissue specimens, 18 included salivary gland tissue. Damage in the form of sialadenitis and other degenerative changes in salivary glands was shown in 4 of the 18 specimens. A consistent pat- tern for chronic sialadenitis was not found among any of the age groups. The authors did not specify the other degenerative changes. However, four patients, ages 21,25,50, and 60 years, demonstrated either a mild, moderate, or severe salivary gland fibrosis. The most severe salivary gland fibrosis was found in the 21-year-old subject who was considered a short-term smokeless tobacco user; a definition for short-term user was not provided. Unlike the findings of Hirsch, Heyden, and ThiIander (18), salivary gland fibrosis or changes were not related to the stage (degree) of the clinical lesion. The authors concluded that there is no doubt that salivary gland fibrosis can be shown and that it is likely to be related to the damage from smokeless tobacco. They also commented that "It is likely that the degree of salivary gland fibrosis and degenera- tive change, along with sialadenitis, may be a factor that is associated with tobacco brand rather than with a generalized reaction caused by alI tobam. ' ' Included among the many questions concerning the effects of smoke less tobacco use on the salivary glands is that of changes on the flow and buffering capacity of saliva. In a sample of 48 Finnish snuff users ages 17 to 21 years (mean 18.9), the resting and stimulated salivary flow was measured (21) (table 1). The subjects refrained from the use of snuff for 1 hour before collection of saliva. The saliva of 10 nonusers was simiIarly collected. The statistically significant findings demonstrated a higher resting salivary flow of snuff users compared with controls. Although the stimulated salivary flow was also higher among the snuff users than the controls, this difference was not statistically significant. Buffering capacity was the same between the two groups. Although 127 these findings offer additional information regarding the effects of smokeless tobacco on the salivary glands, the clinical significance of these effects has not been systematically assessed, nor have the out- come differences related to the different products. Replication studies of these findings are needed before firm conclusions can be made. In contrast to the effects just cited, Archard et al. were unable to identify lesions or dysfunctions associated with smokeless tobacco use (23) (table 2). These investigators carried out histochemical tests on le sions in the oral cavity that were in close proximity to the salivary glands. These tests revealed no evidence of an mflammatory reaction associated with the glands. The interpretation of data within this general area requires caution. Limited evidence suggests a possible relationship between the use of snuff and damage to the salivary glands. Should this be the case, the loss of salivary gland function can result in the decreased production of saliva and the ultimate loss of a protective buffer for the oral epithelium and the teeth against numerous exogenous factors such as infectious agents, including dental caries. THE EFFECTS OF SMOKELESS TOBACCO USE ON TEETH Background and Definitions This section of the chapter addresses the role of various forms of smokeless tobacco in causing or contributing to diseases or conditions of the teeth. Specific effects that are examined include dental caries, abrasion, erosion, plaque and calculus buildup, and staining. For pur- poses of discussion, definitions are offered for a number of terms that are considered to represent commonly held concepts of diseases and conditions of the teeth as evidenced in the relevant scientific literature. o Dental caries-Clinically detectable cavitation of the coronal or root surfaces of the tooth that is caused by acid demineralization of colonizing bacteria on tooth surfaces. o Abrasion-Clinically evident wear of the coronal portion of teeth either generally or focally that appears excessive for a patient of a given age. This is a mechanical effect that is caused by the action of abrasive substances or objects during normal functioning or by oral habits. o Erosion-Loss of tooth structure that is attributable to a chemical agent. o Plaque-Bacterial-laden, proteinaceous material that is continu- ally deposited in the oral cavity through the proliferation of bac- terial types. 128 o Calculus-A concretion that forms on the coronal and exposed root surfaces of teeth through the calcification of bacterial plaques. o Staining-An extrinsic stain deposit that results in discoloration on tooth surfaces. Dental Caries Evidence for the effects of smokeless tobacco use on the teeth is avail- able from several cross-sectional studies (table l), from a limited number of case reports (table 2), and from a limited number of related investiga- tions of the potential for constituents of smokeless tobacco to serve as predisposing or etiologic factors in the development of dental caries. As previously mentioned, Offenbacher and Weathers reported on the oral soft and hard tissue effects of smokeless tobacco use in a study population that comprised 565 males with a mean age of 13.8 years (9). This population typifies the age group that is commonly described as "the cavity-prone years." Although caries rates expressed as decayed, missing, or filled teeth (DMFT) were higher for smokeless tobacco users without gingivitis than for nonusers without gingivitis, these differ- ences were not statistically significant. However, when DMFI' scores for smokeless tobacco users with gingivitis were compared with scores for nonusers without gingivitis, a significantly higher caries prevalence was found among users. Among students who used both snuff and chewing tobacco, the DMFT score was 6.56 + 0.71. This score is significantly elevated compared with scores of nonuser gingivitis-free students and the nonuser group that had gingivitis. There was a 2.4-fold increase in disease experience. In this study, the presence of gingivitis was presented as a cofactor with smokeless tobacco use in the increased prevalence of dental caries. This finding has not bean reported elsewhere, and the biologic explanation is unclear. The different that were noted in caries rates could not be accounted for based upon differences in oral hygiene or the frequency of dental visits-two factors that could potentially affect DMFT scores. The ex- aminers had no knowledge from the self-reported survey forms of the history of smokeless tobacco use among the group that was examined; thus, a degree of study "blindness" was attained. Absolute blindness in these types of surveys is difficult because it is likely that some evidence of smokeless tobacco use (e.g., tobacco residues, stain, odor, and soft tis- sue effects) is observable. No quantifiable doseresponse effect for smokeless tobacco use and dental caries was reported in this study. Dental caries is highly age dependent, and no age adjustment was made in the statistical analysis. A cross-sectional study by Greer and Poulson of 1,119 teenage smokeless tobacco users and nonusers from urban Colorado demon- strated neither "tobaccoassociated dental caries" nor occlusal or in- cisal abrasion of the teeth (7). This finding is not surprising because 129 abrasive effects are cumulative and would likely require a number of years to become evident. The abrasion that has been reported in smoke less tobacco users has been in adults who have used smokeless tobacco products, generally leaf and plug forms of tobacco, for years (10,13). The Greer and Poulson study reported a single case of cervical erosion on the mandibular central incisors. Some case reports have implied a causative role for smokeless tobac- co in the development of dental caries (38,39), while others have postu- lated a potential protective effect from caries (13,40). The presumed mode of protection would be through a greatly increased salivary flow that may provide a buffering action. Additionally, there is evidence that various forms of smokeless tobacco contain fluoride, from a few tenths to several parts per million, which may offer some cariostatic protection (41). At the same time, various types of smokeless tobacco contain up to five different forms of caries-promoting sugars (42). Rvo studies reported that constituents in smokeless tobacco products either cause a proliferation of caries-producing bacteria in vitro or, at the least, do not inhibit bacterial growth in vitro (43,44). The fluoride and sugar contents of smokeless tobacco vary by product type (41). This may explain the in- consistent and equivocal results obtained by different investigators. Variations in reported caries rates, if truly reflective of the larger population of smokeless tobacco users, may represent the clinical out- come of a number of antagonistic or synergistic factors that operate while smokeless tobacco is used. Other Hard Tissue Effects Plaque, calculus, and staining are extrinsic factors that may be asso &ted with smokeless tobacco use. This is clinically important because dental plaque and calculus that is coated with plaque harbor bacteria that can produce acids and toxins and thus bring about dental caries and diseases of the periodontal structures. The stainfng of teeth, restor- ations, and prosthetic appliances have been described as resulting from smokeless tobacco use (13,22,45,46). Van Wyk also reported a constant finding of chronic mflammation of tooth pulps that were extracted from oral snuff users (22). He attributed this as being "probably due to the irritation of the snuff overlying the exposed dentine and cementum." No quantifiable evidence currently documents the risk of smokeless tobacco use compared with nonuse in the development of plaque, calcu- lus, or staining or the relationship of staining to oral disease conditions. CONCLUSIONS 1. Smokeless tobacco use is responsible for the development of a portion of oral leukoplakias in both teenage and adult users. The degree to which the use of smokeless tobacco affects the oral hard 130 and soft tissues is variable depending on the site of action, type of smokeless tobacco product used, frequency and duration of use, predisposing factors, cofactors (such as smoking or concomitant gingival disease), and other factors not yet determined. 2. Dose response effects have been noted by a number of investiga- tors. Longer use of smokeless tobacco results in a higher preva- lence of leukoplakic lesions. Oral leukoplakias are commonly found at the site of tobacco placement. 3. Some snuff-induced oral leukoplakic lesions have been noted upon continued smokeless tobacco use to undergo transforma- tion to a dysplastic state. A portion of these dysplastic lesions can further develop into carcinomas of either a verrucous or squamous cell variety. 4. Recent studies of the effects of smokeless tobacco use on gingival and periodontal tissues have resulted in equivocal findings. While gingival recession is a common outcome from use, gingivitis may or may not occur. Because longitudinal data are not available, the role of smokeless tobacco in the development and progression of gingivitis or periodontitis has not been confirmed. 5. Evidence concerning the effects of smokeless tobacco use on the salivary glands is inconclusive. 6. Negative health effects on the teeth from smokeless tobacco use are suspected but unconfirmed. Present evidence, albeit sparse, suggests that the combination of smokeless tobacco use in individ- uals with existing gingivitis may increase the prevalence of dental caries compared with nonusers without concomitant gingivitis. F&ports of tooth abrasion or staining have not been substantiated through controlled studies; only case reports are available. RESEARCHNEEDS The review of the literature for this component of the report has iden- tified the need for research in each of the areas discussed: the oral soft tissues, the periodontium, the salivary glands, and the teeth. Basically, the effects of the various types and forms of smokeless tobacco in all age groups should be investigated. Controlled studies and comparisons between users and nonusers of smokeless tobacco are needed. Estab lished criteria for assessing tissue changes and disease presence should be applied to permit comparability between studies. Studies should include the identification and control of variables that also may affect these tissues. Such variables may include alcohol use, diet, oral hygiene practices, microbial flora changes, and salivary flow rate, composition, and pH. In addition to these variables, consideration should be given to the effects of concurrent disease states. For example, 131 the effects of smokeless tobacco on dental caries in the presence or absence of gingivitis should be investigated. The natural history of smokeless tobacc&nduced lesions resulting from continued, intermittent, and discontinued smokeless tobacco use needs investigation. Histopathologic evaluations and clinical examina- tions to determine the natural history of oral leukoplakia/mucosal pathology and salivary gland pathology are desirable to understand completely the extent and severity of smokeless tobacco oral effects. In general, incidence and prevalence studies should be implemented, Prospective study designs should be pursued to dssess the temporal relationship between smokeless tobacco use and various health effects. In addition, dose-response studies are needed to assess dose in terms of both duration of use (in months and years) and daily exposure (in minutes and hours). REFERENCES (1) World Health Organization Collaborating Centre for Oral Precancerous Lesions. Definition of leukoplakia and related lesions: An aid to studies on oral precancer. Oral Surg. 46: 518-539,1978. (2) A&II, T., Hohnstrup, P., Kramer, I.R.H., Pindborg, J.J., and Shear, M. International Seminar on Oral Leukoplakia and Associated Lesions Related to Tobacco Habits. Community Dent. Oral Epidemiol. 12: 145154, 1984. (3) International Agency for Research on Cancer. Tobacco habits other than smoking Bet&quid and areca-nut chewing and some related nitro samines. IARC Monogr. Eval. Carcinog. Risk Chem. Hum37: 113,1985. (4) Moore, G.E., Bissinger, L.L., and ProehI, E.C. Tobacco and intraoral cancer. Surg. Forum 3: 685688, 1952. (5) Peacock, E.E., Jr., et al. The effect of snuff and tobacco on the produc- tion of oral carcinoma: An experimental and epidemiological study. Ann. Surg. 151: 542-549, 1960. (6) Smith, J.F., Mincer, H.A., Hopkins, K.P., and Bell, J. Snuff-dipper's lesion. A cytological and pathological study in a large population. Arch. Otokuyngol. 92: 450-456, 1970. (7) Greer, R.O., Jr., and PouIson, T.C. Oral tissue alterations associated with the use of smokeless tobacco by teenagers. Oral Surg. 56: 275~284,1983. (8) Poulson, T.C.. Lindenmuth, J.E., and Greer, R.O., Jr. A comparison of the use of smokeless tobacco in rural and urban teenagers. CA 34: 248-261, 1984. (9) Offenbacher. S., and Weathers, D.R. Effects of smokeless tobacco on the periodontal, mucosal and caries status of adolescent males. J. Oral Pathol. 14: 169-181. 1985. (10) Christen, A.G., Armstrong, W.R., and McDaniel, R.K. Intraoral leukoplakia, abrasion, periodontal breakdown, and tooth loss in a snuff dipper. J. Am. Dent. Assoc. 98: 584-586, 1979. 132 (11) Christen, A.G., McDaniel, R.K., and Doran. J.E. Snuff dipping and tobacco chewing in a group of Texas college athletes. Tex. Dent. J. 97: 6-10, 1979. (12) Hog-e, H.W., and Kirkham, D.B. Clinical management and soft tissue reconstruction of periodontal damage resulting from habitual use of snuff. J. Am. Dent. Assoc. 107: 744-745, 1983. (13) Zitterbart, P.A.. Marlin, D.C., and Christen A.G. Dental and oral effects observed in a long-term tobacco chewer: Case report. J. Indian Dent. Assoc. 62: 17-18, 1983. (14) Axell, T., Momstad. H., and Sundstrom, B. The relation of the clinical picture to the histopathology of snuff dipper's lesions in a Swedish population. J. Oral Pathol. 5: 229-236, 1976. (15) Pindborg, J.J., and Renstrup, G. Studies in oral leukoplakias, II. Effect of snuff on oral epithelium. Acta Derm. Venereol. 4.X: 271-276, 1963. (16) Pindborg, J.J., and Poulsen, H.E. Studies in oral leukoplakias, I. The influence of snuff upon the connective tissue of the oral mucosa. Preliminary report. Acta Pathol. Microbial. Immunol. Stand. 55: 412-414, 1962. (17) Axell, T. A prevalence study of oral mucosal lesions in an adult Swedish population. Odontol. Rev. (Suppl. 36) 27: l-103, 1976. (18) Hirsch, J.M., Heyden, G., and Thilander, H. A clinical histomorphologi- cal and histochemical study on snuff-induced lesions of varying sever- ity. J. Oral Pathol. 11: 387-398, 1982. (19) Frithiof, L., Anneroth. G., Lasson, U., and Sederholm, C. The snuff- induced lesion. A clinical and morphological study of a Swedish material. Acta Odontol. Stand. 41: 53-64, 1983. (20) Greer, R.O., Poulson, T.C., Boone, M.E., Lindenmuth, J., and Crosby, L.K. Smokeless tobacco associated oral changes in the juvenile, adult, and geriatric patients: Clinical and histomorphologic features including light microscopic, immunocytochemical and ultrastructural findings. Gerodontics 2: 3, 1986. (21) Jungell, P., and Malmstrom, M. Snuff-induced lesions in Finnish recruits. Stand. J. Dent. Res. 93: 442-447. 1985. (22) Van Wyk, C.W., The oral lesion caused by snuff. A clinics-pathological study. Medical Proceedings II: 531-537, 1966. (23) Archard, H.O., and Thrpley, T.M.. Jr. Clinicopathologic and histo- chemical characterization of submucosal deposits in snuff dipper's keratosis. J. Oral Pathol. 1: 3-11, 1972. (24) Smith, J.F. Snuff-dippers lesion. A ten-year follow-up. Arch. Otolaryngol. 101:276-277, 1975. (25) Brightman, V.J. Laboratory procedures. In: Malcolm A. Lynch (ed). Burket's Oral Medicine-Diagnosis and `Beatment. Philadelphia, J.B. Lippincott Company, 1977, pp. 723-724. (26) Mehta, F.S., Gupta, P.C., and Pindborg, J.J. Chewing and smoking habits in relation to precancer and oral cancer. J. Cancer Res. Clin. Oncol. 99 35-39, 1981. 133 (27) `Qldesley, W.R. Tobacco chewing in English coahniners (2). Malignant transformation in a tobacco-induced leukoplakia. Br. J. Oral Surg. 14: 93-94, 1976. (2.9) Roed-Petersen, B, and Pindborg, J.J. A study of Danish snuff-induced oral leukoplakia. J. Oral Pathol. 2: 301-313, 1973. (29) Vogler, W.R., Lloyd, J.W., and Mihnore, B.K. A retrospective study of etiological factors in cancer of the mouth, pharynx, and larynx. Cancer 15: 246-258, 1962. (30) McGuirt, W.F. Snuff dipper's carcinoma. Arch. Otolaryngol. 109: 757-760, 1983. (31) Sundstrom, B., Momstad, H.. and AxelI, T. Oral carcinomas associated with snuff dipping. Some clinical and histological characteristics of 23 tumours in Swedish males. J. Oral Pathol. 11: 245-251,1982. (32) Rosenfeld, L., and Cahaway, J. Snuff dipper's cancer. Am. J. Surg. 106: 840-844, 1963. (33) Landy, J.J., and White, H.J. BuccogingivaI carcinoma of snuff dippers. Am. Surg. 27: 442-447,196l. (34) Banndczy, J., and Sugar, L. Progressive and regressive changes in Hungarian oral leukoplakias in the course of longitudinal studies. Com- munity Dent. Oral Epidemiol. 3: 194-197, 1975. (35) Pindborg, J.J., Reibel, J., and Hohnstrup, P. Subjectivity in evaluating oral epithehal dysplasia, carcinoma in situ and initial carcinoma. J. Oral Pathol. 14: 698-708. 1985. (36) Mod&, T., Lavstedt, S., and Ahlund, C. Relation between tobacco con- sumption and oral health in Swedish schoolchildren. Acta Odontol. Stand. 38: 223-227, 1980. (37) Hecht, S.S., Chen. C.B., Hirota, N., Omaf, R.M., and `Iho, T.C. Tobacco- specific nitrosamines: Formation from nicotine in vitro and during tobacco curing and carcinogenicity in strain A mice. J. Natl. Cancer Inst. 60: 819-824, 1978. (38) Sitzes, L. On chewing tobacco. ADA News 8: 2, 1981. (39) Croft, L. Smokeless tobacco: A case report. Tex. Dent. J. 99: 15-16, 1981. (40) Shannon, I.L., and `RodahI, J.N. Sugars and fluoride in chewing tobac- co and snuff. Tex. Dent. J. 96: 6-9, 1981. (41) Going, R.E., Hsu, S.C., Pollack, R.L., and Haugh, L.D. Sugar and fluoride content of various forms of tobacco. J. Am. Dent. Assoc. 100: 27-33, 1980. (42) Hsu, SC., Pollack, R.L., Hsu, A.F., and Going, R.E. Sugars present in tobacco extracts. J. Am. Dent. Assoc. 101: 915-918, 1980. (43) Lindemeyer. R.G., Baum, R.H., Hsu, S.C., and Going, R.E. In vitro ef- fect of tobacco on the growth of oral cariogenic streptococci. J. Am. Dent. Assoc. 103: 719-722. 1981. 134 (44) Falkler, W.A., and Zimmerman, M.L. Effect of smokeless tobacco ex- tracts on the growth of streptococcus mutans. Presented at the Annual Meeting of the International Association of Dental Research, Las Vegas, Nevada, March 1985. (45) Christen, A.G. The case against smokeless tobacco: Five facts for the health professional to consider. J. Am. Dent. Assoc. 101: 464-469,198O. (46J U.S. Department of Health and Human Services. Draft report to the Surgeon General by the Inspector General on smokeless tobacco. December 20, 1985. (47) Waldron, C.A., and Shafer, W.G. Leukoplakia revisited. A clinicopatho logic study of 3,256 oral leukoplakias. Cancer 36: 1386-1392, 1975. (48) Waldron, C.A., and Shafer, W.G. Current concepts of leukoplakia. Int. Dent. J. 10: 350-367, 1960. (49) Shafer, W.A., Hine, M.K., and Levy, B.M. Oral Pathology, ed. 2. Philadelphia, W.B. Saunders Co., 1969, pp. 80-81, 85. /So) Liie, H.. and Silness, J. Periodontal disease in pregnancy. I. Prevalence and severity. Acta Odontol. Stand. 21: 533-551. 1963. 135 Chapter 4. NlCOTlNE EXPOSURF' PHARMACOKINmCS, ADDICTION, AND OTHER PHYSIOLOGIC EFFECTS CONTENTS Introduction .,,...................................141 Pharmacokinetics of Nicotine Levels of Nicotine in Smokeless Tobacco Absorption of Nicotine Distribution of Nicotine Nicotine Elimination Nicotine and Cotinine Levels in Users of Smokeless Tobacco Time Course of Nicotine `Bunover During Daily lbbacco Use References 141 141 141 142 142 143 143 143 Nicotine Addiction Associated With Smokeless lbbacco Use Background and Definitions Commonahties Between Ibbacco Use and Other Addictive Substances Experimental Studies of the Abuse Liability and Physical Dependence Potential of Nicotine Evidence That Orally Delivered Nicotine (Including Via Smokeless lbbacco) Has a Liability for Abuse and a Potential to Produce Physical Dependence References....................................... 144 144 146 157 166 168 Physiologic and Pathogenic Effects of Nicotineand Smokeless l'Uxcco Physiologic Effects of Nicotine Nicotine, Smokeless Tobacco, and Human Diseases . NonnicotineRelated Adverse Metabolic Consequences References 175 175 176 179 ,179 Conclusions . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . ...182 ResearchNeeds....................................184 139 This chqter examin es the consequences of exposure to nicotine from smokeless tobacco. It draws from the vast literature on the effects of nicotine delivered via smoking and intravenously and includes recent evidence of the effects of orally delivered nicotine. The first section describes the pharmacokinetics of nicotine, includ- ing absorption, distribution, and elimination. The data presented indi- cate that nicotine is present in smokeless tobacco in significant amounts and that users attain blood levels of nicotine similar to those produced by cigarette smoking. The second section reviews the established evidence that nicotine is an addictive and dependenceproducing substance, having a number of important characteristics in common with prototypic addictive and dependenceproducing substances, as well as substantial experimental evidence of its abuse liability and dependence potential. Given the nice tine content of smokeless tobacco, its ability to produce high and sus- tained blood levels of nicotine, and the well-established data implicating nicotine as an addictive substance, one may deduce that smokeless tobacco is capable of producing addiction in users. In addition, very re- cent studies provide direct confirmation that nicotine delivered orally from smokeless tobacco and nicotine chewing gum is addictive, produc- ing abuse liability and dependence potential. The final section of the chapter reviews the multisystem physiologic effects of nicotine and examin es the evidence pertaining to the potential contributory role of nicotine in the causation of several diseases. PHARMACOKINETICS OF NICOTINE Levels of Nicotine in Smokeless Tobacco `Ibbacco is a plant product, and therefore differences exist in nicotine content among and within different strains of tobacco. Nicotine content among smokeless tobacco products also differs: moist snuff contains 4.56 to 15.1 mg nicotine per gram (1); plug tobacco has been measured to contain 17.2 mg per gram (2). Assuming a daily consumption of 10 grams of smokeless tobacco, the habitual user can be exposed to roughly 130 to 250 mg nicotine per day, of which varying amounts may be absorbed. By comparison, cigarette tobacco averages 15 mg nicotine per gram or 9 mg nicotine per cigarette (3). A person who smokes a pack of cigarettes per day therefore can be exposed to 180 mg nicotine per day. Absorption of Nicotine Nicotine is a weak base (pKa 7.9). In its ionized form, as in the acidic environment of most cigarette smoke, nicotine crosses membranes poorly. As a consequence, there is virtually no buccal absorption of nice 141 tine from cigarette smoke. In contrast, smokeless tobacco products are buffered to an alkaline pH that facilitates absorption. The rate of absorption of nicotine from smokeless tobacco depends on the product and the route of administration. With fineground nasal snuff, blood levels of nicotine rise almost as fast as those that are observed after cigarette smoking (4). The rate of nicotine absorption with the use of oral snuff (and presumably chewing tobacco) is more gradual (5). People who use oral smokeless tobacco, particularly those who chew tobacco, generate large amounts of saliva, some of which is expecto rated and some of which is swallowed. Due to first pass metabolism in the liver following absorption from the intestines, the bioavailability of swallowed nicotine is approximately 30 percent (6). By changing how much is chewed, how much is held inside the mouth, and how much saliva is expectorated or swallowed, the user of smokeless tobacco has considerable control over the dose of nicotine that is absorbed. Distribution of Nicotine Smoking is a unique form of drug administration in that entry into the circulation is through the pulmonary rather than the portal or sys- temic venous circulations. The lag time between smoking and the appearance of nicotine in the brain is even shorter than after intrave- nous injection. Nicotine enters the brain quickly, but then brain levels decline rapidly as it is distributed to other body tissues. The rapid brain uptake of nicotine from smoking allows easy puff-topuff titration of desired nicotine effects and partly may explain the highly addictive nature of cigarette smoking. In contrast, the concentrations of nicotine that enter the brain from smokeless tobacco use are likely to be lower (6), and the pharmacologic ef- fects may differ. The rate of exposure to psychoactive drugs is an impor- tant determinan t of their effects. Thus there could be differences in the ef- fects of nicotine that is taken by smoking compared to using smokeless tobacco, even with the same average body concentrations of nicotine. Nicotine Elimination Nicotine is rapidly and extensively metabolized primarily in the liver but also to a small extent in the lung and kidney. Renal excretion depends on urinary pH and urine flow and accounts for 2 to 35 percent of total elimination (78). The half-life of nicotine averages 2 hours, although there is considerable individual variability that ranges from 1 to 4 hours (9). The major metabolites of nicotine are cotinine and nicotineN-oxide. Neither metabolite appears to be pharmacologically active (8). Because of its long half-life, cotinine is commonly used as a marker of nicotine intake in survey and cessation studies. It should be recognized, however, that first pass metabolism of swallowed nicotine 142 may result in wtinine levels that are disproportionately higher than nicotine levels with the use of smokeless tobacco compared to the use of cigarettes. Nicotine and Cotinine Levels in Users of Smokeless Tobacco Blood or plasma concentrations of nicotine in cigarette smokers who were sampled in the afternoon generally ranged from 10 to 50 ng/ml (10). The increment in blood nicotine concentration after a single cigarette is smoked ranges from 5 to 30 ng/ml, depending on how the cigarette is smoked (llJ2). In users of moist oral snuff or chewing tobacco, the levels of nicotine increase an average from 2.9 to 21.6 ngiml during 8 hours of repeated use (1). In habitual users of nasal snuff, blood levels of nicotine increased on average by 12.6 ng/ml after a single dose of snuff, and levels aver- aged 36 nglml after multiple doses (4). Similarly, blood cotinine concen- trations averaged 197 ng/ml and 411 ng/ml in groups of oral and nasal tobacco users, respectively, compared to an average cotinine level of 300 q/ml for cigarette smokers described in many studies 11,4). These comparisons indicate that the intake of nicotine and nicotine levels in habitual users of smokeless tobacco are similar to those that are ob- served in habitual cigarette smokers. Time Course of Nicotine Turnover During Daily Tobacco Use Tobacco use is commonly considered to be a process of intermittent dosing of nicotine, which in turn is rapidly eliminated from the body. Smoking produces considerable variations from highest to lowest blood nicotine levels from one cigarette to the next cigarette. However, con- sistent with a half-life of 2 hours, nicotine accumulates over 6 to 8 hours of regular smoking, and nicotine levels persist overnight, even as the smoker sleeps (13). The same accumulation is probable with repeated smokeless tobacco use. Thus as with the smoker, the smokeless tobacco user may be exposed to nicotine for 24 hours each day. References (1) Hoffmann, D., Harley, N.H., Fisenne, I., Adams, J.D., and Brunne mann, K.D. Carcinogenic agents in snuff. JNCI 76: 435-437,1986. (2) Hoffmann, D., Hecht, S.S., Omaf, R.M., Wynder, E.L., and Tso, T.C. Chemical studies on tobacco smoke. XLII. Nitrosonornicotine: Presence in tobacco, formation and carcinogenicity. In: E.A. Walker, P. Bogovski, and L. Griciute (eds.). Environmental N-Nitroso Com- pounds. Analysis and Formation (IARC Scientific Publications No. 14). Lyon, France, International Agency for Research on Cancer, 1976, pp. 307-320. 143 (3) Benowitz, N.L., Hall, S.M., H eming, R.I., Jacob, P., III, Jones, R.T., and Osman, A.L. Smokers of low-yield cigarettes do not consume less nicotine. N. Engl. J. Med. 309: 139-142, 1983. (4) Russell, M.A.H., Jarvis, M.J., Devitt, G., and Feyerabend, C. Nicotine intake by snuff users. Br. Med. J. 283: 814-817, 1981. (5) Russell, M.A.H., Jan&, M.J., West, R.J., and Feyerabend, C. Buccal absorption of nicotine from smokeless tobacco sachets. Lancet 8468: 1370, 1985. (6) Jenner, P., Gorrod. J.W., and Beckett, A.H. The absorptionof nicotine 1 `-N-oxide and its reduction in the gastrointestinal tract in man. Xeno- biotica 3: 341-349, 1973. (7) Beckett, A.H., Gorrod, J.W., and Jenner, P. A possible relation be tween pKa and lipid solubility and the amounts excreted in urine of some tobacco alkaloids given to man. J. Pharm. Pharmacol. 24: 115-120, 1972. (8) Benowitz, N.L., Kuyt, F., Jacob, P.. Jones, R.T., and Osman, A-L. Coti- nine disposition and effects. Clin. Pharmacol. Ther. .%5? 139142,1983. (9) Benowitz, N.L., Jacob, P., III, Jones, R.T., and Rosenberg, J. Inter- individual variability in the metabolism and cardiovascular effects of nicotine in man. J. Pharmacol. Exp. Ther. 221: 368-372,1982. (10) Russell, M.A.H., Jarvis, M., Iyer, R., and Feyerabend, C. Relationship of nicotine yield of cigarettes to blood nicotine level concentration in smokers. Br. Med. J. 280: 972-976, 1980. (11) Armitage, A.K., Dollery, C.T., George, C.F., Houseman, T.H., Lewis, B.J., and timer, D.M. Absorption and metabolism of nicotine from cigarettes. Br. Med. J. 4: 313-316, 1975. (12) Herning, R.I.. Jones, R.T., Benowitz, N.L.. and Mines, A.H. How a cigarette is smoked determines nicotine blood levels. Clin. Pharmacol. Ther. 33 84-91, 1983. (13) Benowitz, N.L., Kuyt, F., and Jacob, P., III. Circadian blood nicotine concentration during cigarette smoking. Clin. Pharmacol. Ther. 32: 758-764. 1982. NICOTINE ADDICTION ASSOCIATED WITH SMOKELESS TOBACCO USE Background and Definitions Clinical observations and data, historical anecdotes, and sworn testi- mony all support the conclusion that some users of smokeless tobacco are unable to abstain permanently from smokeless tobacco, even when iIl health is apparent (1). Such observations suggest that smokeless tobacco use can become a form of drug addiction or dependence.* o The terms "addictnn and dependence" will be used almost interchangeably throughout this section While man argue the value of one of these terms over the other, it is im i-tam to note that in the context of this chapter ???? B f& B dress the questkon of whether ruwtine resulting from smo or smokeless tobacco use leads an individual to lose voluntary control over bk or her use of t&acco products (i.e.. does the drug cause either dependence or addiction). 144 This section of the report will evaluate the scientific evidence that smokeless tobacco is an addictive substance whose use results in drug dependence. Drug dependence as used in this review is defined in accor- dance with the World Health Organization's Expert Committee on Drug Dependence (2) and other recognized sources (3). Drug dependence is substanceseeking behavior that is controlled by the activity of a con- stituent drug in the central nervous system and displaces other behavior such that drug seeking assumes greater priority. IUzrance and physiologic withdrawal may or may not be present (23). and the severity of dependence may vary considerably among individuals. The scientific standard for classifying a drug as likely to cause addic- tion or dependence is based on the degree to which "abuse liability" and "physical dependence potential" are present. Both terms are accepted terminology of the Committee on Problems of Drug Dependence and the Addiction Research Center (ARC) of the National Institute on Drug Abuse (4,5F and are commonly accepted to refer to drugs whose actions are mediated by the central nervous system Abuse liability refers to drug effects that contribute to compulsive self-administration, often in the face of excessive financial cost, physical and social dysfunction, and the exclusion of more socially acceptable behaviors (56). Physical dependence potenti (also referred to as physiological dependence potential) pert&s to the direct physiologic effects that are produced by the repeated administration of a drug that results in neuroadaptation (34). Neuroadaptation is characterized by demonstrated tolerance to the effects of the drug and the occurrence of physiologic withdrawal signs following the termination of drug administration. Physiologic or physical dependence, as evidenced by physiologic and behavioral rebound (withdrawal) effects, is neither necessary nor suffi- cient to define drug dependence (35). Nevertheless, the process of drug dependence and abuse entails physical components, including physical interactions between drug and tissue in the central nervous system (specific receptors in the case of some drugs such as nicotine and opioids) that are critical~ Three lines of evidence are important to assess the abuse liability and physical dependence potential of smokeless tobacco use. The first in- volves inference from the systematic comparison of tobacco use (includ- ing smokeless forms) to the use of prototypic dependenceproducing drugs (e.g., alcohol, morphine, and cocaine) to determine whether the t A concept that is central to many discussions of drug de 8" dence is that the substance reduces damage or debilitation. This asp& of tob.scco dependence will not be a dressed here because extensive 8. ta already exist in- dicnting the actual toxicity of tobacco and there is widespread recognition even by tobacco users that the sub stance I.3 harmful. 145 patterns of tobacco use, as well as the behavioral and physiologic effects of such use, are similar to those of the prototypic dependenceproducing drugs. This issue is discussed below in the section entitled "Commonali- ties Between Tobacco Use and Other Dependence-Producing Substances." The second line of evidence emerges from recent studies in which nicotine was evaluated using the same methods and criteria that have been used to evaluate any substance that is suspected of causing abuse and physical dependence. This deductive approach evaluates whether nicotine meets rigorous experimental criteria as a drug that has sub stantive liability for abuse and physical dependence potential. This issue is discussed in the section entitled "Experimental Studies of the Abuse Liability and Dependence Potential of Nicotine." The third line of evidence comes from recently completed studies that involve direct assessments of the abuse liability and dependence poten- tial of orally given nicotine. E xamination of these studies provides indi- cations of whether the consumption of nicotine through oral forms of administration delivers pharmacologically active quantities of nicotine to the bloodstream and whether smokeless tobacco itself meets specific criteria for abuse liability and dependence potential. This issue is dis- cussed in the section entitled "Evidence That Orally Delivered Nicotine (Including Smokeless Tobacco) Has a Liability for Abuse and a Poten tial to Produce Dependence. " Taken together, the first and second lines of evidence support the con- clusion that smokeless tobacco contains an addictive substance. The third line of evidence suggests that delivery of the addictive substance (nicotine) in the form of smokeless tobacco does not alter its addictive properties. Commonalities Between Tobacco Use and Other Addictive Substances The assertion that tobacco use can occur as a form of drug addiction rests firmly on the observed commonalities between the use and effects of tobacco and the use and effects of addictive substances such as alto hoi, opium, and coca. Systematic reviews of these commonalities have been published (%ll), and the major points that tobacco and addictive substances have in common are as follows: o A centrally (CNS) active substance (drug) is delivered. o Discriminative (subjective) effects are centrally mediated. o The substance (drug) is a reinforcer for animals. o The patterns of acquisition and maintenance of substance inges- tion are orderly. o The patterns of self-administration of the substance are orderly. 146 o The patterns of self-administration of the substance vary as a func- tion of the dose that is consumed. o Tolerance to the behavioral and physiologic effects of the sub stance develops with repeated use (neuroadaptation). o Therapeutic effects may be produced by the substance. o The treatment of addiction resulting from the substance (drug) involves similar strategies. The evidence concerning tobacco and these factors is presented in the following subsections. `Ibbacco Use Delivers a Centrally Active Substance-Nicotine The fundamental commonality between tobacco use and the use of known addictive substances is the delivery of a chemical to the central nervous system The primary agent in tobacco, nicotine, is delivered to the central nervous system in all commonly used forms of tobacco (12). The fact that cigarette smokers will substitute smokeless tobacco, when cigarettes are not available or when the use of combustibles is restricted, certainly suggests that different forms of tobacco use pro duce acceptably similar effects for the user (13). . . . Dmnmmative Effects of Nicotine Are Centrally Mediated Nicotine, like other drugs of abuse, produces doserelated effects in animals, which an be attenuated by centrally acting antagonists (1416). When the animals confuse these effects with other drugs (i.e., effects partially generalize to other drugs of abuse), it is more likely to be a drug like amphetamine rather than a sedativelike drug (17). These findings are also consistent with data derived from studies with humans in which the doserelated effects of intravenously given nicotine were attenuated by mecamylamine pretreatment (18). Nicotine Is a Reinforcer for Animals Most drugs that are abused by humans are voluntarily self- administered when they are made available to animals in laboratory studies; in other words, the drug serves as a reinforcer or a reward (19,20). Such findings confirm that the physiologic effects of the drug in the central nervous system are sufficient for the substance to control behavior by virtue of its reinforcing effects. Definitive studies ???? were undertaken in the early 1980's support this statement. As seen in table 1, nicotine has now been shown to function as a reinforcer for five non- human animal species and under a variety of conditions (21,227. F'urther- more, its functional behavioral effects are similar to those engendered when other drugs of abuse (e.g., cocaine) serve as reinforcers. Patterns of Acquisition and Maintenance of Tobacco Use Are Orderly The use of tobacco, like that of prototypic addictive substances, is often initiated due to peer influences (2.5). The contribution of social 147 TABLE I.-Summary of Reports in Which Nicotine Was Available Under Intravenous Drug Self-Administration Procedures Study SpC!CieS Reinforcement Schedule Main Finding Comment Deneau Rhesus and Inoki Monkey (19371 Yanagita, Rhesus Ando, Monkey OiIlUIIUl, and Ishida (1974) Lang. Hooded Latiff. Rat McQueen. and Singer (1977) Singer, Hooded Simpson, Rat andLang (1973) Fixed-ratio 1 (FR 1). Several doses of nicotine were tested. Experiment 1: FR 1. Several doses of nicotine and lefetamine and saline were t&MI. Nicotine did not - serve as a reinforcer when compared to saline or lefetamine. Experiment 2: Stable rates of No direct test of FR 1. Several nicotine S-A reinforcing doses of nicotine occurred in most efficacy was done. were continuously subjects but were available for at not clearly related least 4 weeks. to dose. Experiment 3: Progressive ratio (PR) procedures. `ho doses of nicotine and saline and three doses of cocaine were teSti. At 0.2 mg/kg nice Nicotine was tine, response marginaNy rein- rates slightly forcing when exceeded those compared to maintained by cocaine. dine or the lowest cocaine dose (0.03 mg/kg). FR 1. Nicotine and saline were tested in food- sated and food- deprived rats. Concurrent [(FR 1: nicotine). (Fixed- time 1 min.: food pellet)] in food- deprived rats. Subsequently, the rats were food- sated. Two monkeys Currently initiated self- accepted criteria administration to assess reinforc- (S-A); the others ing efficacy were required a prim- not achieved. ing procedure. In food-deprived - (but not food- sated) rats, nicotine was a reinforcer when compared to saline. Food satiation Results were simi- decreased rate of lar to those nicotine S-A, how- obtained when ever, nicotine was rats were similarly a reinforcer in tested with both conditions. ethanol. 148 TABLE l.-Continued Study Species Reinforcement Schedule Main Finding Comment Griffiths, Baboon Brady, and Bradford (1979) Hanson, Albino Rat Iveskr, and Moreton (1979) Latiff, Hooded Smith, and Rat Smith and Hooded L=% Rat (1980) Goldberg, Squirrel Speahnan, Monkey and Goldberg (1981) FR 160 followed by 3-hr. timeout. Several doses of nicotine and saline were substituted for cocaine. FR 1. Several doses of nicotine and saline were teSti. Cone (FR 1: injec- tion) (FT 1 min.: food pellet). Several doses of nicotine and saline were teSti. FR 1. One dose of nicotine and saline were tested. Second order schedule FI 1 or 2 min. (FR 10: stimulus) followed by 3-min. timeout. One dose of nico- tine and saline was tested. Number of nice- tine injections per day did not exceed that of saline. Mecamylamine (centrally acting antagonist) but not pent&km (peripherally act- ing antagonist) altered S-A behavior. Nicotine was a reinforcer relative to saline. Urine pH manipulations had mild effects on rate of S-A only during initial exposure to nicotine. Nicotine was established as a reinforcer both with and without a concurrent food delivery schedule Caffeine, ephedrine, and a variety of other similarly tested stimulants did serve as rein- forcers relative to saline in this paradigm. Group data suggest that nicotine was a reinforcer; however, there was no clear dose effect curve. Rate of S-A was inversely related to dose during initial exposure to nicotine but not after nicotine S-A was established. - in food-deprived but not food-sated rats. Nicotine main- Demonstrated the tained high rates importance of of responding. ancillary environ- Rates decreased mental stimuli in markedly when (1) maintaining high saline replaced rates of nicotine, (2) the responding. brief stimuh were omitted, and (3) subjects were pretreated with mecamylamine. 149 TABLE l.-Continued Study Species Reinforcement Schedule Main Finding Comment Ator and Baboon Griffiths (1981) Dougherty, Rhesus Miller. - Monkey Todd, and Kosten- bauder (1981) Goldberg and Spealman (1982) Singer, Wallace, and HalI (19821 Squirrel Monkey Long Evans Rat FR 2 followed by Nicotine was 15-sec. timeout. marginally rein- Several doses of forcing compared nicotine and saline to saline across a and cocaine were narrow dose tested. range. FI 16 and second order FI 1 min. (FR 4: stimulus). Several doses of nicotine and saline were tested. FI 5 min. Several doses of nicotine and cocaine and saline were tested. CONC [FR 1: nicotine) (FT 1 min.: food pellet)]. One dose of nicotine was tf?Skd. Nicotine main- tained higher rates of S-A than saline under the FI and second order schedules but was only a margjnally effec- tive reinforcer when continu- ously available. Nicotine and Initial dose response curve was inverted U-shaped, and final dose response curve was flat (from abstract of study). Establishment of nicotine as a rain- forcer required several months using procedures that typically require only a few days to establish cocaine or codeine as reinforcers. This study also cocaine were quali- showed that tatively similar nicotine could reinforcers when serveasa compared to punisher similar to saline. Cocaine electric shock. maintained higher rates of respond- ing in one of two monkeys. Meca- mylamine pre- treatment reduced rates of nicotine S-A. A group of rats Extended the with 6-OHDA range of lesions in the scheduled-induced nucleus accum- behaviors that are hens S-A nicotine inhibited by such at lower rates lesions. than a sham- lesioned group. 150 TABLE l.-Continued Study Species Reinforcement Schedule Main Finding Comment Spealman and Goldberg (1982) Risner and Goldberg (1983) Henning- field, Miyasato, and Jasinski (1983) Goldberg and Henning field (1983) Squirrel Monkey Beagle Dog Human and Squirrel Monkey Second order FI 1, 2, or 5 min. (FR 10 stimulus) and FI 5 min. schedules were tested. Several doses of nicotine and cocaine and saline were tested. FR 15 followed by 4 min. timeout. Several doses of nicotine, cocaine, and saline were tested. Progres- sive ratio schedule was used. FR 10 followed by 1 min. timeout. Several doses of nicotine and saline were tested. FR 10 followed by 1 min. timeout. Several doses of nicotine and saline were tested. Nicotine and Nicotine's rem- cocaine main- forcing efficacy tained similar was comparable to patterns of that of cocaine. responding on the schedules. Nice tine, but not cocaine S-A, decreased to salinelike rates when animals were pretreated with mecamylamine. Nicotine and Cocaine main- cocaine main- tained substan- tained qualita- tially greater tively similar response rates patterns of than nicotine. responding and were reinforcers relative to saline. Mecamylamine pretreatment reduced nicotine but not cocaine S-A. Number of nice Nicotine produced tine injections subjective effects generally ex- similar to those ceeded number of produced by intra- saline injections venous cocaine and were inversely and had both rein- related to nicotine forcing and dose. Post-session punishing effects. cigarette smoking was suppressed by nicotine. Patterns of In both the responding were human and mon- qualitatively key subjects, similar in both there was evidence species. Number that nicotine func- of nicotine injec- tioned with both tions exceeded reinforcing and number of saline punishing injections in 3 of properties. 4 human and 3 of 4 monkey subjects. 151 support to the initiation of tobacco use may be even greater than with illicit drugs, because family members, other social models, and advertis- ing often tolerate, approve, or promote tobacco use while disapproving the use of some nonprescription drugs (`4). Also, as is the case with addictive drugs, an accelerated pattern of development of tobacco use has been observed, which is followed by relatively stable drug intake. Initially, the level of consumption increases gradually from the first day of use until some point, perhaps several years later, when it becomes relatively stable over time. Although many factors can operate to pro duce such a biphasic pattern of intake, it is generally assumed that tolerance and learnin g factors account for the gradual acceleration and that a level of optimum drug effect combined with toxicity and adverse effects at higher doses takes over to produce the stabilization phenome non. A preliminary survey, conducted at Johns Hopkins University, indicates that nicotine, whether administered as cigarette smoke or smokeless tobacco, does not differ from other drugs in this regard. That is, tobacco users tend to begin smoking a few cigarettes a day or con- sume a portion of a container of smokeless tobacco each day and gradu- ally increase consumption levels over a period of months or even years before they stabilize the amount they finally use (personal communica- tion, J.E. Henningfield). Patterns of Tobacco Self-Administration Are Orderly Daily patterns of cigarette smoking are orderly. Addicted smokers tend to smoke their first cigarette within 30 minutes of waking from a night of sleep and find it difficult to abstain from tobacco use for more than a few hours (2.5). If smoking behavior is relatively unconstrained, regular patterns develop that closely resemble those of psychomotor stimulant self-administration in animals (20). Similar orderly patterns of tobacco self-administration are evident with cigarette smoking by humans. Several studies have demonstrated that across successive puffs on a cigarette, puff duration decreases and interpuff intervals tend to increase (26,27,2-!$29), although these changes are multifactor- ially determined (30). Anecdotal reports by smokeless tobacco users suggest that while consumption patterns are necessarily different (e.g., some keep a plug in their mouth almost continually during their waking hours) they are no less regular and orderly. Tobacco Self-Administration Varies as a Function of Nicotine Dose The effective dose of a substance may be varied by changing the quantity of drug per unit (the unit dose), by pretreating the individual (animal or human) with either an agonist or antagonist, or by altering the rate of elimination of the substance. Studies that involve these three manipulations have been done extensively with other drugs and more recently with nicotine, The results across study, drug, and species are remarkably similar. For general reviews of human and animal studies 152 see Griffiths, Bigelow, and Henningfield (20) and Henning-field, Lukas, and Bigelow (31). See Gritz (32) and Henningfield (33) for recent reviews of the nicotinespecific literature. Over a wide range of dose levels, fre quency of self-administration is inversely related to dose but drug in- take is directly related to dose, reflecting partial compensatory changes (26,32). Pretreatment with other agonists (or forms of nicotine) reduces drug taking, e.g., decreases cigarette smoking, (34) and reduces pre ferred nicotine concentration of tobacco smoke (35). Pretreatment with antagonists initiaIly increases drug self-administration. For example, the centrally and peripherally acting ganglionic blocker, mecamyla- mine, but not the peripherahy acting blocker, pentolinium, increases subsequent smoking rates and increases preferred nicotine concentra- tions of tobacco smoke (36,37). In addition, altering the elimination rate of nicotine alters the amount of nicotine that is self-administered in the form of tobacco smoke (38). There has been debate over the degree to which smokers regulate their nicotine intake, i.e., the "titration" hypothesis. It is now generally agreed that smokers do not precisely titrate their nicotine intake any more than animals titrate their intake of reinforcing drugs (except under extremely limited conditions) or humans titrate their intake of other reinforcing drugs (20). However, when dose manipulations are observed and objective, sensitive dependent variables are measured in both animals and humans (26,32,33), most of the studies demonstrate an increase in smoking as cigarette nicotine content falls below accus- tomed levels and a decrease in smoking when cigarette nicotine content is unusually high (32). Kozlowski and his coworkers describe these find- ings in terms of a "boundry" model of dose compensation (39). Tolerance of Nicotine Develops With Repeated Use (Neuroadaptation) The administration of mostdrugs of abuse results in neuroadaptation as measured by tolerance to the repeated administration of the drug and a subsequent rebound (withdrawal) when drug administration is terminated (3). Tolerance to drug effects is determined either by the diminished response to repeated doses of a drug or the requirement of increasing doses to achieve the same drug effect. Tolerance to the behavioral and physiologic effects of nicotine has been studied for decades (33). As is the case with other drugs of abuse, a variety of mechanisms accounts for tolerance to many of nicotine's effects, includ- ing metabolic (40), behavioral (41#), and physiologic tolerance (44-46). More recently, studies have shown that the effects of nicotine that are suspected to be critical to the addiction process also show tolerance with repeated dosing (4799. Physiologic dependence on drugs is determined by showing that ter- mination of drug administration produces a syndrome of effects that is generally opposite to those produced by drug administration. This syn- drome is reversible, at least in its early stages, by administration of the 153 drug. Prolonged drug abstinence (detoxification) results in ultimate return to baseline (normal) values of behavioral and physiologic frmc- tions. It is now clear that repeated tobacco administration produces physiologic dependence that is specifically due to nicotine administra- tion. Recent data that confirm this fact are reviewed in the section on Dependence Potential of Nicotine. Nicotine Produces Therapeutic Effects Most drugs of abuse have specific therapeutic applications; nicotine is no exception (4850). The degree to which the therapeutic effects of nicotine depend upon the individual's history of nicotine use, as opposed to the possibility that nicotine is efficacious for preexisting conditions, remains to be investigated. Similar issues are true for other drugs of abuse as well. Pomerleau and his coworkers (51) have studied a variety of mechanisms by which the possibly weak, initial reinforcing effects of nicotine can be greatly strengthened by subtle effects on mood, cogni- tion, and normal physiologic and behavioral functioning. For instance, as will be described below, nicotine may produce a small, but important, enhancement of work performance. These effects appear to be mediated by the effects of nicotine on hormonal release and regulation The following is a brief summary of some of the effects of nicotine, con- sidered therapeutic by tobacco users, that have been investigated. Several studies have shown that nicotine enhances performance on a variety of cognitive tasks that involve speed, reaction time, vigilance, and concentration (5265). These effects are strongest in cigarette smokers who are deprived of cigarettes. However, such performance enhancement was also evident after the administration of nicotine to nonsmokers and was produced by increasing the nicotine dose in per- sons who were already smoking. Nicotine may also be a useful mood regulator by virtue of its release of norepinephrine from the adrenal medulla (56). Norepinephrine release is also stimulated by excitement, exercise, sex, antidepressant drugs, and other drugs of abuse, sug- gesting that cigarette smoking may function pharmacologicalIy to alleviate boredom and stress. Finally, as an anoretic (57+X?), nicotine ap- pears to function in three ways: by decreasing the efficiency with which food is metabolized (61,62); by reducing the appetite for foods that con- tain simple carbohydrates (sweets) (63); and by reducing the eating that may occur in times of stress (64). Nicotine may also function as an am& lytic by reducing responsiveness to stressful stimuli and enhancing mood (56). In addition, nicotine reduces aggressive responses in experi- mental situations (65). A well-documented therapeutic role for nicotine as a drug is evident in the treatment of tobacco abstinence for many individuals following dependent patterns of tobacco use, e.g., as assessed by the Fagerstrom Tolerance Questionnaire (25). This test provides both scientific and prac- tical evidence of the role of nicotine in tobacco dependence. It is well 154 established that abstinence from tobacco in heavy cigarette smokers produces signs and symptoms of rebound that can be reversed by resumed tobacco use and at least partially reversed by other forms of nicotine administration (66). For example, nicotine gum treatment for cigarette smoking is efficacious, although a variety of factors limit suc- cess rates &I).* This drug substitution strategy is analogous to those obtained when intravenous opioid users are treated with other opioids given via other routes. For example, methadone administration may reverse signs and symptoms of opioid withdrawal, while leaving the pa- tient feeling partially treated yet likely to relapse if not provided with an adjunctive behavioral treatment (67). Although the euphoriant properties of drugs can stand apart from collateral therapeutic actions (as is the case with morphine, am- phetamine, and alcohol), attention to such drug effects may enhance the efficacy of treatment. Because nicotine, in the form of tobacco, is widely available, is relatively inexpensive, and is in a convenient form for precise dose regulation, it provides an ideal means of self-medication. These effects may contribute to the abuse liability of tobacco and are of demonstrable significance in the treatment of tobacco addiction (51). Sii Strategies Are Involved in the lkeatment of Tobacco Addiction and Other Forms of Drug Addiction If tobacco use is a form of drug addiction, then strategies of treat- ment of other forms of drug addiction should be applicable. Most avail- able information and existing strategies for treatments of tobacco use are based on nonpharmacologic approaches. Such approaches have been no more useful in the treatment of tobacco dependence than in the treatment of dependence of opioids, stimulants, sedatives, or alcohol. On the contrary, experience in the treatment of drug addiction disorders makes clear the importance of addressing the pharmacologic components of the addiction (67). This conclusion is strengthened by the observation that persons being treated for opioid addiction regard tobacco to be as necessary as methadone (68) and that persons success- fully treated for other kinds of drug addiction are unable to give up tobacco (ss/. This provides the support for the fundamental premise that tobacco addiction generally constitutes an independent health- impairing disorder. Specific treatment implications relating to cigarette smoking as a form of drug abuse are considered below. `lb the extent that tobacco use is similar to other forms of drug abuse, treatment strategies that are used for drug abusers may be applied to the treatment of cigarette smoking. Although it is not the purpose of this chapter to describe in detail the treatment for cigarette smoking, a o These therapeutic effects are produced by nicotine chewing gum. an orally administered form of nicotine that is approved by the Food and Dru J Administration (FDA). The tmn only and is commonly u .r m is obtainable in the United States by prescrip by physicians to help indiw uals quit smoking. 155 few commonalities, as well as differences, are worth mentioning. Four basic pharmacologic treatments for drug abuse provide the advantage of licit administration of an agent controlled by a certified clinician These involve substitution therapy (e.g., methadone for opiate depen- dence) in which a more manageable form of the drug is provided accord- ing to a prearranged maintenance protocol; blockade therapy (e.g., naltrexone for opiate dependence) in which the effects of the abused drug are blocked by pretreatment with an antagonist; and nonspecific supportive therapy in which the patient is treated symptomatically, ex- emplified by the temporary use of bemodiazepines during alcohol detoxification (67). All three approaches have been used in the treat- ment of cigarette smoking with varying degrees of success (48). A fourth strategy of pretreating the patient with a drug that results in adverse side effects when the subsequent abused drug is taken (e.g., treatment of alcoholism with disuhiram) has not been systematically explored with tobacco. The most recent, widely used treatment for cigarette smoking, and the first of those recognized as efficacious by the FDA, is modeled directly after the treatment of heroin addiction by methadone substitu- tion. This treatment is nicotine gum substitution 170). It is a practical application of the postulate that tobacco use is basically a form of drug addiction on nicotine. This recognition is especially relevant here, because smokeless tobacco is an oral form of nicotine. All of the relevant therapeutic data support the premise that compulsive tobacco use en- tails nicotine addiction, which in the form of tobacco exposes the user to health hazards, and that therapeutic strategies paralleling those for other forms of drug abuse are effective in treatment. Differences appear to be principally related to the social tolerance of tobacco addiction, relative to other forms of drug addiction, which contribute to greater difficulty in treating this form of drug abuse. Summary of Commonalities Between Tobacco and Prototypic Addictive Drugs The preceding review has shown that tobacco shares many points in common with prototypic addictive drugs. These similarities provide a strong conceptual basis for the categorization of tobacco as an addictive drug. The behavioral process is orderly, tobacco self-administration results in the delivery of a centrally active drug (nicotine), and the drug appears to be the major dete rminant in the control of the compulsive behavior of tobacco self-administration These findings are consistent with those expected with animal and human subjects, as determined across a broad range of studies of drugs of abuse (.). In summary, tobacco, opium, and coca produce different effects but share a number of important similarities. Whereas large doses of opioids can produce a debilitating sedation, high doses of coca alkaloids (cocaine HCI) produce levels of behavioral excitation that are not nor- 156 mally produced by tobacco; but the intake of all of these substances leads to compulsive use. Compulsive use and the other commonalities described in the preceding subsections provide compelling evidence that tobacco use can be a form of drug dependence or addiction. The next major question is what element(s) of tobacco are critical to control- ling the behavior of the user. The conceptual leap from habitual behavior to drug abuse and addiction can be made only on the basis of evidence that a specific psychoactive drug is critical to the behavior. The next section on the abuse liability and dependence potential of nicotine will address this question. Experimental Studies of the Abuse Liability and Physical Dependence Potential of Nicotine The comparison of tobacco to prototypic addictive drugs is the basis for concluding that compulsive tobacco use is a form of drug dependence behavior in which nicotine plays an important role. `lb test this hypothesis further, it should be possible to show that nicotine is an abusable substance even in the absence of the many stimuli associated with cigarette smoking. This can be done by evaluating nicotine in ac- cordance with methods and criteria that have been used to assess any substance that is suspected of causing abuse and physical dependence. Onehalf century of research at the NIDA Addiction Research Center, and research in other laboratories, has produced valid and reliable ex- perimental methods to evaluate a substance's potential to cause abuse and to produce physical dependence. The methods are empirically based on generally accepted examples of drug addiction, most notably opioid dependence (e.g., morphine) and, to a lesser degree, psychomotor stimulant dependence (e.g., cocaine) and sedative dependence (e.g., bar- biturates and alcohol). These methods encompass standards for assess- ing the two dimensions of drug addiction-abuse liability and physical dependence potential. The evidence that is related to the abuse liability and physical dependence potential of nicotine is presented below. Abuse Liability of Nicotine Abuse liability refers to drug effects that contribute to compulsive self-administration, often in the face of excessive financial cost, physical and social dysfunction, and the exclusion of more socially acceptable behaviors (S,S). In other words, it entails those effects of a substance that contribute to diminution of voluntary control over the use of the substance by the individual. Objective methods to assess abuse liability are available and have been used to assess diverse agents 1'). These methods have been readily adapted to studies of nicotine abuse liability, with consideration given to the fact that nicotine has more rapid effects than many other drugs of abuse. 157 The hypothesis is that nicotine is psychoactive and serves as a euphoriant and reinforcer. Psychoactivity and euphoria are determined by assessing the pharmacodynamic subjective effects of single doses of the drug ("singledose" or "abuse liability" studies) and are validated by observed behavioral and physiologic responses. Reinforcing efficacy is determined by assessing the ability of the drug to strengthen and maintain orderly patterns of behavior when the subject is permitted ac- cess to the drug (i.e., the prototypic "self-administration" study). Phxunuzcodynamic Effects of Nicotine. In human studies of nicotine related psychoactivity, volunteers are given a range of doses of the test compound and placebo under doubleblind conditions. Persons with histories of drug abuse are used because they can accurately disuiminate compounds with a potential for abuse and can compare the effects of the compounds to those of abuse drugs (5). In one study, three doses of nicotine were given both intravenously and in the form of tobacco smoke under controlled conditions (71). Nicotine produced a similar profile of ef- fects (figure 1). Self-reported (subjective), observer-reported (behavioral), and physiologic variables were measured before, during, and after drug administration In brief, nicotine was shown to be psychoactive, as evidenced by the reliable disc rimination of nicotine from placebo, Self- reported effects of nicotine peaked within 1 minute after administration (by either route) and dissipated within a few minutes: peak and duration of response were directly related to the dose. The two hallmark indicators of euphoria in such studies are the Lik- ing Scale (Single Dose Questionnaire) and the Morphine Benzedrine Group (MBG) Scale (Addiction Research Center Inventory [ARCI]) (5). Responses on the 5-point Liking Scale, which asked how much the drug was liked (0 = "not at all," 4 = "an awful lot") are presented in figure 2. Nicotine produced responses on the Liking Scale similar to those of morphine and d-amphetamine. MBG Scale scores of the ARC1 were con- sistent with the Liking Scale data, confirming that nicotine, given by both routes of administration, was a euphoriant. In another comparison between drugs, subjects more frequently identified nicotine injections as cocaine. Similar results for intravenous and inhaled nicotine were also obtained on several physiologic measures, including pupil diameter, blood pressure, and skin temperature. These data confirmed that nicotine, given in either tobacco smoke or intravenously, was the critical pharma- cologic compound accounting for these effects of tobacco smoke. A sub- sequent study showed that nicotine's subjective and physiologic effects could be partially blocked by pretreating the subjects with the antago- nist mecamylamine (18). Results of studies with animals also indicate that nicotine produces discriminable effects, and the data suggest that animals identify nicotine as being more similar to cocaine than to placebo or pentobarbital, but not identical to cocaine (17). 158 FIGURE l.-This figure is a summary of the data from a study of the liability of nicotine delivered as tobacco smoke (filled symbols-IN or intravenous injections (open symbols-IV). Dose is presented on the hori- zontal axes. Even with a controlled smoking procedure, nicotine dose administration via cigarette smoke is more variable (producing flatter doseresponse functions) than when given intravenously. Also, important effects of nicotine are covert though reliable and orderly (e.g., relaxed feel- ings, symptom scores). The finding that a low dose of tobacco smoke was more effective in reducing desire to smoke than a low dose of intravenous nicotine is consistent with the fact that satisfaction from smoking is also due to stimuli provided by the cigarette and the smoke. Self-Administration ofNicotine. The second abuse liability dimension uses the "self-administration" procedure to examine the conditions under which a subject will voluntarily take the drug. Self-administration studies determine whether the drug serves as a biologically effective, positive reinforcer (or reward). Variants of these strategies are con- ducted in both animal and human subjects, thereby providing a means of establishing the biologic generality of the phenomena, while control- ling the possible confounding influence of personality, social, or cultural variables. A high degree of concordance between findings from animal FIGURE Z.-This figure presents data from a series of abuse liability studies conducted at the Addiction Rezxmch Center. The rmdings that Liking Scale scores are directly related to dose and exceed placebo values are important in identifying dependenceproducing drugs. Intra- venous nicotine produced the same elevated dose-response function as highly addictive narcotics (e.g., morphine) and a prototypic stimulant (d-amphetamine). These data are also consistent with the lower abuse liability of chlordiazepoxide and almost negligible abuse liability of zomepirac. Administration of intravenous cocaine results in a function similar to that shown for intravenous nicotine, except that the cocaine dose levels must be increased by a factor of 5 to 10. MORPHINE (SC) 2 t / :: P 7.5 IS 30 W a z 2 PENTOBARBITAL a W = I o P 120 240 BUPRENORPHINE (SL) / 0 1 P P .75 I.5 3 CHLORDIAZEPOXIDE (PO) 0 4 I 1 1 I P 50 100 200 DRUG DOSE (mgl PENTAZOCINE (lrn) P:T t A-9-THC (PO) t: P 5 IO 20 ZOYEPIRAC (PO) P 200 400 800 and human studies has been established over a wide range of drugs (20). Therefore, this section focuses on the results of studies using human volunteers. The methods developed in animal studies can be used to assess whether the pharmacologic activity of a drug maintains self-administra- 160 FIGURE 3.-This figure shows the patterns of nicotine self- administration that occurred when volunteer cigarette smokers were given the opportunity to take injections of nicotine, but not smoke cigarettes, during 3-hour tests. The amount of nicotine available was roughly comparable to that obtained by smoking cigarettes. The sub jects smoked less following sessions in which they took nicotine than following sessions in which only saline (the placebo) was available. I.V. NICOTINE INJECTIONS SUBJECT &!/kg BE I I I I I I I I 27 KOI I I I I I 27 SKw I I I I I L22 KUI I I I I II III I I I 11 I22 PEw I 11 I I I I II I II II I I I 18 LA1 I I I I I I I I I I I I I II I II I I I II 18 KE 11 111 11 1 I I I 1 II I I I I I I II I 11 1 13 -3 HOURS-I tion paralleling drug seeking and drug taking by individuals in the natural environment or "real world." The strategy is particularly useful in studies of nicotine, because it precludes confounding by other stimuli that are associated with tobacco smoke inhalation (e.g., the tobacco brand, smell of the smoke, and lighting-up rituals). In one such study, tobaccodeprived volunteers were tested during 3-hour sessions in which 90 presses on a lever resulted in either a nice tine or placebo injection (72). All six subject% voluntarily self- administered nicotine (figure 3). Patterns of self-administration (injec- tions) were similar to those observed when human subjects smoke cigarettes and when rhesus monkeys take intravenous amphetamine in- jections in comparable experimental situations (20). One subject, who lacked a history of drug abuse, exhibited an acquisi- tion pattern of nicotine self-administration that developed gradually over several sessions. The pattern was a prototypic example of drug 161 abuse development. Doubleblind substitution of saline for nicotine resulted in cessation of the self-injection behavior of subject KO (figure 3). Subjects who were given access to both nicotine and placebo concur- rently (by pressing alternate levers) chose nicotine, confirm@ that nicotine had come to serve as a positive reinforcer (73). These data indi- cate that the pharmacologic activity of nicotine was critical to the maintenance of the behavior. Nicotine self-administration has been studied in a variety of non- human species under a variety of experimental conditions (74). As noted earlier, recent results confirm that nicotine can function as an effective reinforcer although the conditions under which it serves as a reinforcer for animals are more restricted than those for morphine or cocaine (21). Nicotine self-administration via cigarette smoke or smokeless tobacco may provide ideal confluences of conditions for the establishment and maintenance of nicotine dependence in humans (33) with the presence of immediate and abundant peripheral taste and olfactory stimuli (75). Implications of Pharmucodynamic and Self-Administration Studies. The results of the pharmacodynamic and self-administration studies provide direct evidence that nicotine itself, and apart from its beii pre sented in combination with all of the orosensory properties of tobacco smoke, is an abusable drug. That is, nicotine meets the criteria of being psychoactive: it serves as a euphoriant and as a reinforcer. These find- ings strongly suggest that nicotine parallels other drugs (e.g., morphine in opium use, cocaine in coca leaf use, and ethanol in alcoholic beverage consumption) in its ability to maintain self-administration. The find- ings are of sufficient strength that the relevant public health implica- tions have already been incorporated into issues of public health policy by the former Director of the National Institute on Drug Abuse, Dr. W. Pollin (76), the U.S. Public Health Service (77), and the former Secretary of the Department of Health and Human Services, Mrs. M. Heckler (78). Physical Dependence Potential of Nicotine Physical dependence potential (also referred to as physiological dependence potential) pertains to the direct physiologic effects that are produced by the repeated administration of a drug that results in neuro adaptation (3,4). Neuroadaptation is characterized by demonstrated tolerance to the effects of the drug and the occurrence of physiologic withdrawal signs following the termination of drug administration. Physical dependence potential studies are conducted according to standardized tests, using methods such as the substitution approach in which an active drug is removed and replaced with either a placebo or another form of the drug (5). Although many studies on the effects of tobacco abstinence on mood, behavior, and physiologic functions have been conducted, until recently, the classic "direct addiction" or "substitution" methodologies had not been used to study the physical dependence potential of nicotine (79). 162 The absence of such studies and the fact that many critical markers of tobacco abstinence are not overt or easily measured (e.g., change in affect, EEG, and cognitive performance impairment) have led to ques- tions about the severity of the tobacco withdrawal syndrome (33). However, as shown below, abstinence from chronic tobacco or oral nice tine use is followed by a syndrome of behavioral and physiologic changes that are orderly, replicable, specific to nicotine, and of func- tional consequence in relapse to tobacco following abstinence. The apparent absence of withdrawal symptoms among some people is not inconsistent with the finding that nicotine has the potential to produce physical dependence. As is true for users of opiates (e.g., heroin), the magnitude of the withdrawal syndrome is related to a variety of factors such as dosage and individual predispositions (80). Definition of lbbacco Withdrawal There are abundant data indicat- ing neuroadaptation to tobacco use, showing that this adaptation is at least partially nicotine specific and that termination of chronic tobacco use produces a behavioral and physiologic rebound or withdrawal syn- drome (33). This has been stated in the Diagnostic and Statistical Manual (DSM) of the American Psychiatric Association (APA) as follows (81): Tobacco Withdrawal (APA, DSM, III, 1980). The essential feature is a characteristic withdrawal syndrome due to recent cessation of or reduction in tobacco use that has been at least moderate in duration and amount. The syndrome includes craving for tobacco, irritability, anxiety, difficulty concentrating, restlessness, headache, drowsiness, and gastrointestinal disturbances. It is assumed that this syndrome is caused by nicotine withdrawal, since nicotine is the major pharma- cologically active ingredient in tobacco. Withdrawal does not occur with all smokers; but in many heavy cigarette smokers, changes in mood and performance that are prob ably related to withdrawal can be detected within two hours after the last cigarette. The sense of craving appears to reach a peak within the first 24 hours after the last cigarette, thereafter gradually declining over a few days to several weeks. In any given case it is difficult to dis- tinguish between a withdrawal effect and the emergence of pychologi- cal traits that were suppressed, controlled, or altered by the effects of nicotine. This definition by the American Psychiatric Association represents a reasonable consensus from various reviews of the literature on cigarette smoking and physiologic dependence on tobacco (3,13,32,82&3). It is based on experimental data and clinical observations from cigarette smoking treatment studies demonstrating that certain signs and symp- toms are of unusualIy high prevalence during the first few days of absti- nence. Decreased heart rate and blood pressure have been studied experimentally (84), as well as changes in cortical EEG potentials (85,86,), 163 changes in urine catecholamine excretion (83, and weight gain (57). Other possible concomitants of tobacco withdrawal reported clinically include headaches, gastrointestinal disturbances, insomnia, and fatigue (82,87). A variety of behavioral effects occurs when tobacco or nicotine administration is abruptly Wmina ted in human and animal subjects, including increased irritability, aggressiveness, and anxiety; perfor- mance also is impaired in various psychomotor and learning tests such as simulated driving, vigilance, and paired-associate learning (88-W). Self-reported desire to smoke cigarettes ("craving") increases sharply for about 1 day following abstinence, then gradually declines over the course of about 1 week to a lesser level (91). Most of these signs and symptoms of withdrawal subside over 1 to 2 weebs; however, some former tobacco users report that the desire to smoke may recur for many years and may be evoked by specific environmental stimuli that were previously associated with smoking, such as after meals or in selected social situa- tions. This, too, parallels the powerful conditioning phenomena that are reported to be associated with other drugs of abuse (92). Evidence of lbbacco Withdrawal Symptoms. There is compehing evi- dence that acute tobacco abstinence produces a rebound (withdrawal) syndrome. This evidence comes from studies of two laboratories in which increases in low-i?equency EEG bands and decreases in cortical activity were observed during the first day of tobacco abstinence (&QX$. These effects were immediately reversed when the subjects were allowed to smoke two cigarettes. In a study of self-reported withdrawal symptomatology, 40 partici- pants completed four 25&m questionnaire forms daily for 2 weeks (93). Subjects were divided into two groups: totally abstinent and partiaky abstinent whose smoking levels were maintained at an average of 60 per- cent. Four symptom clusters emerged: (1) drowsiness in both groups declined over the first week and then increased over the second week, forming a U-shaped function; (2) physical symptoms (e.g., headaches and gastrointestinal disturbances) in both groups declined rapidly the first week and then remained stable across the second week; (3) psychological symptoms (e.g., anxiety and irritability) in both groups paralleled physi- cal symptoms; and (4) craving symptoms in the totally abstinent group closely paralleled physical and psychological symptoms, whereas craving levels of the partially abstinent subjects remained elevated across the 2 weeks. The finding that partial abstinence is accompanied by persistent craving symptomatology is similar to the results of studies on the treat- ment of illicit opioid dependence with methadone. In these studies, low- dose methadone maintenance is associated with a persistent opioid crav- ing (94). An important series of studies on the dependence potential of nicotine has recently been completed at the University of Minnesota (95,%$97). The goals of these studies were to determine reliable and valid indicators 164 of tobacco withdrawal by examining physical, subjective, and behav- ioral reactions to tobacco deprivation. The first three studies of this series evaluated the dependence potential of tobacco and established a reliable battery of measures. In a residential study, 27 smokers resided for 7 days on a research ward ($5). Following baseline, they were assigned to abstain from smoking or to continue smoking for 4 days. Physiologic, subjective, and behavioral measures were obtained and analyzed. The second study was conducted on a nonresidential basis to assess tobacco withdrawal in the nonlaboratory environment (W. In this study, signs and symptoms of tobacco withdrawal were measured in 100 smokers. Following baseline measurements, subjects were randomly assigned to either nicotine or placebo gum, to be chewed at each subject's own rate. The subjects returned on three different occasions for assessment. The third study assessed the reliability of the tobacco withdrawal syndrome within subjects ($7). This study employed a modified, within-subject ex- perimental design; baseline smoking, tobacco deprivation, return to baseline smoking, and tobacco deprivation were assessed in each sub- ject. The results of all three studies demonstrated that the syndrome of withdrawal that occurs reliably and consistently in chronic smokers after tobacco deprivation includes decreased heart rate, increased caloric intake/eating, an increased number of awakenings during sleep, an increased desire to smoke cigarettes, and increased confusion. Other changes that were found, but not consistently, included increased irri- tability and decreased vigor. A prospective examination of data from both residential and nonresidential studies revealed that there were no statisticahy significant differences between men and women in either number or severity of tobacco withdrawal symptoms (98. A subsequent study was designed to assess the relationship between tobacco withdrawal symptoms and pre and post-cigarette blood nice tine levels, prec&arette cotinine levels, change in nicotine level from pre to post-cigarette, half-life of nicotine, and total smoke exposure ($9). Twenty subjects were required to smoke cigarettes for 3 days using a portable recorder that allowed measurements of smoking topography in a nonlaboratory environment. Blood samples were drawn to deter- mine blood nicotine and cotinine levels. Subjects abstained from cigarettes for the next 4 days. A battery of tests to measure tobacco withdrawal symptoms was administered. In general, results showed an inconsistent relationship between measures of nicotine intake and tobacco withdrawal. The most consistent finding was the relationship of the desire to smoke cigarettes to blood nicotine and cotinine levels and change in nicotine from pre and post-cigarette; that is, the higher the nicotine and cotinine level and "nicotine boost," the greater the desire for cigarettes during abstinence. The three initial studies that were conducted at the University of Minnesota (95,,9697) systematically examined the physiologic depen- 165 dence produced by chronic tobacco use. This work represents a major advance in furthering the understanding of tobacco dependence. The NIDA Addiction Research Center is also nearing the completion of a series of studies on the physical dependence potential of tobacco and the degree to which oral nicotine treats the abstinence syndrome. F're- liminary data analysis confirms the findings from the Minnesota studies. Implications of Physical Dependence Potential Studies. These recent studies confirm and extend the findings of earlier investigations that demonstrated that nicotine had the potential to produce. physiologic dependence. It is now known that the syndrome is orderly and is due to the administration and withdrawal of nicotine. The overt signs are more subtle than those marking opioid and sedative withdrawal, but these signs are not necessarily less important to the individual For instance, withdrawal effects such as mood changes, performance deficits, and weight gain may be of considerable importance to the normal function- ing of the individual. It is anticipated that just as detoxification and treatment of opioid and sedative dependence have benefited from im- proved understanding of these syndromes of withdrawal, so also may detoxification and treatment of tobacco withdrawal benefit. Evidence That Orally Delivered Nicotine (Including Via Smokeless Tobacco) Has a Liability for Abuse and a Potential to Produce Physical Dependence As previously indicated, moist snuff contains as much as 15.1 mg nicotine per gram; plug tobacco contains 17.2 mg per gram (100,101). Lower-nicotinecontaining brands exist. However, marketing efforts encourage (and users demonstrate) graduation to the higher-nicotine containing products (1). These levels of nicotine are substantial, since the relative potency of nicotine is 5 to 10 times greater than that of co caine in producing di suiminable subjective effects (1 to 2 mg of nicotine given intravenously, orally, or inhaled produces reliable behavioral and physiologic effects). `Ityo studies have confirmed that typical patterns of smokeless tobac- co use result in the delivery of quantities of nicotine that produce plasma nicotine elevations comparable to those produced when ciga- rettes are smoked (102,1031. These studies also found that smokeless tobacco use reflected several of the indices of abuse liability and physical dependence potential. Smokeless tobacco users self- administered substantial quantities of nicotine; the patterns of smokeless tobacco use were orderly and stable; and subjective and behavioral effects may be produced from such use. More recently, a new form of smokeless tobacco, moist brown tobacco in tea bag-like pouches, was also shown to deliver pharmacologically active quantities of nicotine to the central nervous system (104). 166 Reinforcing Properties of Nicotine in the Form of Chewing Gum There is growing evidence that nicotine is reinforcing and has the potential to produce dependence even when absorbed through the buc- cal mucosa (and therefore more slowly) via chewing gum (nicotine pola- crilex). One recently completed study involved the self-administration of either a nicotine or placebo-containing chewing gum by smokers who had quit smoking (105). When given a choice between placebo and nicotine chewing gum, subjects preferred nicotine to placebo and self- administered the nicotine gum throughout each day.* These data are particularly compelling, because nicotine, in the form of the nicotine polacrilex, is in an ion-bound complex. In this preparation, the nicotine is released and absorbed slowly compared to the nicotine in smokeless tobacco; and the polacrilex form of nicotine administration appears to be of relatively low abuse liability. This study also demonstrated that instructions by a physician can alter patterns of gum use and preference (105). These data, which suggest that instructions can modulate the self- administration of orally delivered nicotine, are in keeping with the well- known fact that physicians control their patients' use of narcotics, sedatives, and stimulants. Physical Dependence Potential of Smokeless Tobacco Hatsukami and coworkers, at the University of Minnesota, studied neuroadaptation (physiologic dependence) in smokeless tobacco users (10s). All 16 subjects in the study used moist snuff and no other nicotine-delivering product. Measures of mood, feeling, behavior, and physiologic function were cornpar& 1.t baseline and during abstinence. Subjects showed significant signs and symptoms of nicotine with- drawal as measured by decreased resting pulse, attenuated orthostatic pulse changes, and increases in tobacco seeking ("craving"), eating, sleep disruptions, and confusion. A study with nicotine g-urn showed orally delivered nicotine may cause physical dependence (107). The subjects that were tested had been treated for tobacco dependence with nicotine gum that they used on a daily basis for at least 1 month. Eight subjects were then tested over the course of 4 weeks. They were given nicotine-containing gum during the first and fourth weeks; during the second and third weeks, they received nicotine gum for 1 week and placebo gum for the other. During the week that placebo gum was presented, seven subjects showed signs and symptoms of withdrawal, and two subjects relapsed to smoking or nicotine-containing gum. This study confirms that oralIy given nicotine has the potential to produce physical dependence. These findings were most recently confirmed by another study that showed development of physical dependence to nicotine gum in patients treated for tobacco dependence (1 Ch9). * Selfadmimstratron rook place ar an average raw of 7 4 pieces compared to an average of 1.2 paces of placebo gum per day. 167 References (1) ConnoIIy, G.N.. Winn, D.M., Hecht, S.S.. Henningfield, J.E., Hoffmann, D., and Walker, B. Science public policy and the reemergence of smoke less tobacco. N. Engl. J. Med. (in press). (2) World Health Organization. Technical Report Series, No. 407. Geneva, Switzerland, 1969. (3) Jaffe, J.H. Drug addiction and drug abuse. In: A.G. Gihnan, L.S. Goodman T.W. RaII, and F. Mm-ad @Is.). Goodman and Gihnan's Pharmacological Basis of Therapeutics. New York, Macrnihan, 1985, pp. 532-581. (4) Brady, J.V., and Lukas, S.E. (eds.). The Committee on Problems of Drug Dependence, Inc. Testing Drugs for Physical Dependence Potential and Abuse Liability (NIDA Research Monograph 52). Washington, D.C., U.S. Government Printing Office, 1984. (5) Jasinski, D.R., Johnson, R.E., and Henningfield, J.E. Abuse liability assessment in human subjects. Trends in PharmacologicaI Sciences 5: 196-200, 1984. (6) Jasinski, D.R. Assessment of the abuse potentiality of morphinelike drugs (methods used iu man). In: W.R. Martin (ed.). Handbook of Experi- mental Pharmacology, Vol. 45. Drug Addiction I. Berlin, West Germany, Springer-VerIag. 1977, pp. 197-258. (7) Jarvik, M. The role of nicotine in the smoking habit. In: W.A. Hunt (ed.). Leaning Mechanisms in Smoking. Chicago, AkIine, 1970, pp. 155-190. (8) Russell, M.A.H. Cigarette smoking National history of a dependence disorder. Br. J. Med. PsychoI. 44: l-16, 1971. (9) Jarvik, M. Further observations on nicotine as the reinforcing agent in smoking. In: W.L. Dunn (ed.). Smoking Behavior: Motives and Incen- tives. Washington, D.C., Winston, 1973, pp. 3349. (10) Jaffe, J.H., and Kanzler, M. Smoking as an addictive disorder. In: N.A. Krasnegor (ed.). Cigarette Smoking as a Dependence Process (NIDA Research Monograph 23). Washington, D.C., U.S. Government Printing Office, 1979, pp. 4-23. (11) Henningfield, J.E.. Griffiths, R.R., and Jasinski, D.R. Humau depen- dence on tobacco and opioids: Common factors. In: T. Thompson and C.E. Johanson (eds.). Behavioral Pharmacology of Human Drug Dependence (NIDA Research Monograph). Washington, D.C., U.S. Government Printing Office, 198 1. (12) Schmiterlaw, C.G., Hansson. E., Andersson, G., Appelgren, L.E., and Hoffman, PC. Distribution of nicotine in the central nervous system Ann. N.Y. Acad. Sci. 143: 2-14, 1967. (13) Russell, M.A.H. Tobacco smoking and nicotine dependence. In: R.J. Gibbons, Y. Israel, H. KaIant, R.E. Popham, W. Schmidt, and R.G. Smart (eds.). Research Advances in Alcohol and Drug Problems. New York, Wiley, 1976, pp. l-46. (14) FImemam, J.A. Nicotine as a discriminative stimuIus to behavior: Its char- acterization and relevance to smoking behavior. In: N.A. Krasnegor (ed.). Cigarette Smoking as a Dependence Process (NIDA Research Monograph 23). Washington, DC., U.S. Government Printing Office, 1979, pp. 5869. 168 (15) Stolennan, I.P. Disc riminative stimulus properties in nicotine: Correla- tions with nicotine binding. proceedings of the International Symposium on Tobacco Smoking and Health: A Neurobiologic Approach (in press). (16) Rosecrans, J.A., and Meltzer, L.T. Central sites and mechanisms of ac- tion of nicotine. Neurosci. Biobehav. Rev. 5: 497-501, 1981. (17) Stolerman, I.P., Pratt, J.A., Garcha, H.S.. Giardini, V., and Kumar. R. Nicotine cue in rats analyzed with drugs acting on cholinergic and 5-hydroxtryptamine mechanisms. Neuropharmacology 22 1029-1033, 1983. (18) Henning-field, J.E., Miyasato, K., Johnson, R.E., and Jansinski, D.R. Rapid physiologic effects of nicotine in humans and selective blockade of behavioral effects by mecamylamine. In: L.S. Harris (ed.). Problems of Drug Dependence, 1982 (NIDA Research Monograph 43). Washing- ton, D.C., U.S. Government Printing Office, 1983, pp. 259-265. (19) Griffiths, R.R., and Balster, R.L. Opioids: Similarity between evalua- tions of subjective effects and animal self-administration results. Clin. Pharmacol. Ther. 25: 611-617,1979. (20) Griffiths, R.R., Bigelow, G.E., and Henning-field, J.E. Similarities in animal and human drug taking behavior. In: N.K. Mello (ed.). Advances in Substance Abuse: Behavioral and Biological Research. Greenwich, Connecticut. JAI Press, 1980, pp. l-90. (21) Henningfield, J.E.. and Goldberg, S R. Nicotine as a reinforcer in human subjects and laboratory animals. Pharmacol. B&hem. Behav. 19989-992,1983. (22) Goldberg, S.R. Nicotine as a reinforcer in animals. In: M.E. Jarvik (ed.). Nicotine and Appetite. Proceedings of the International Symposium on Tobacco Smoking and Health: A Neurobiological Approach (in press). (23) U.S. Department of Health and Human Services, Public Health Service. The Health Consequences of Smoking for Women: A Report of the Sur- geon GeneraL Washington, D.C., U.S. Government Printing Office, 1980. (ZQ) Haertzen, C.A., Kocher, T.R., and Miyasato, K. Reinforcement from the first drug experience can predict later drug habits and/or addiction: Results with caffeine, cigarettes, alcohol. barbiturates, minor and ma- jor tranquilizers, stimulants, marijuana, hallucinogens, heroin, opiates and cocaine. Drug Alcohol Depend. 11: 147-165,1983. (25) Fagerstrom, K. Measuring degree of physical dependence to tobacco smoking with reference to individualization to treatment. Addict. Behav. 3: 235-241, 1978. (26) Griffiths, R.R., and Henningfield, J.E. Pharmacology of cigarette smoking behavior. `Bends in Pharmaceutical Science 3: 260-263,1982. (27) Chait, L.D., and Griffiths, R.R. Smoking behavior and tobacco smoke intake: Response of smokers to shortened cigarettes. Clin. Pharmacol. Ther. 32: 90-97, 1982. (28) NemethCoslett, R., and Griffiths, R.R. Determinants of puff duration in cigarette smokers: I. Pharmacol. Biochem. Behav. 20: 965-971, 1984. (29) NemethCoslett, R., and Griffiths, R.R. Determinants of puff duration in cigarette smokers: II. Pharmacol. Biochem. Behav. 21: 903-912.1984. 169 (a) NemethCoslett, R., and Griffiths, R.R. Effects of cigarette rod length on puff volume and carbon monoxide delivery in cigarette smokers. Drug Alcohol Depend. 15: 1-13, 1985. (31) Henningfield, J.E., L&as, S.E., and Bigelow, G.E. Human studies of drugs as reinforcers. In: S.R. Goldberg and I.P. Stolerman (eds.). Behavioral Analysis of Drug Dependence. New York, Academic Press, 1986, pp. 69122. (32) Grit-z, E.R. Smoking behavior and tobacco abuse. In: N.K. Mello (ed.). Advances in Substance Abuse. Greenwich, Connecticut, JAI Press, 1980, pp. 91-158. (33) Henning-field, J.E. Behavioral pharmacology of cigarette smoking. In: T. Thompson, T.B. Dews, and J.E. Barrett (eds.). Advances in Behavioral Pharmacology, Vol. IV. New York, Academic Press, 1984, pp. 131-210. (34) Grabowski, J., and Hall, S.M. Pharmacological adjuncts in smoking cessation (NIDA Research Monograph 53). Washington, D.C., U.S. Government Printing Office, 1985. (Z) Rose, J.E., Herskovic, J.E., Trilling, Y., and Jarvik, M.E. `Ransdermal nicotine reduces cigarette craving and nicotine preference. Clin. Phar- macol. Ther. 38: 450-456, 1985. (55) Stolerman, I.P. Goldfarb, T., Fink, R., and Jarvik, M.E. Influencing cigarette smoking with nicotine antagonists. PsychopharmacologiaZ& 247-259, 1973. (37) Nemeth-Coslett, R., Henningfield, J.E., O'Keeffe, M.K., and Griffiths, R.R. Effects of mecamylamine on cigarette smoking and subjective ef- fects. Psychopharmacology 86: 420-425,1986. (38) Benowitz, N.L., and Jacob, P.. III. Nicotine renal excretion rate influ- ences nicotine intake during cigarette smoking. J. Pharmacol. Exp. Ther. 234: 153-155, 1985. (39) Kozlowski, L., and Herman, C.P. Controlled tobacco use. In: W. Harding and N. Zinberg (eds.). Control Over Intoxicant Use: Pharma- cological, Psychological and Social Considerations. New York, Human Sciences Press, 1982, p. 207. (40) Beckett, A.H., and `Riggs, E.J. Enzyme induction in man caused by smoking. Nature 216: 587,1967. (41) Clarke, P.B.S.. and Kumar, R. The effects of nicotine on locomotor activity in non-tolerant and tolerant rats. Br. J. Pharmacol. 78: 324337, 1983. (42) Stitzer, M., Morrison, J., and Domino, E.F. Effects of nicotine on fixed- interval behavior and their modification by cholinergic antagonists. J. Pharmacol. Exp. Ther. 171: 166-177, 1970. (43) Stolerman, I.P., Bunker, P., and Jarvik, M.E. Nicotine tolerance in rats: Role of dose and dose interval. Psychopharmacology 34: 317-324,1974. (44) Faulkerbom, Y., Larsson, C., and Nordberg, A. Chronic nicotine expo- sure in rats: A behavioral and biochemical study of tolerance. Drug Alcohol Depend. 8: 51-60,1981. 170 (4%) Domino, E.F. Behavioral, electrophysiological, endocrine and skeletal muscle actions of nicotine and tobacco smoking. In: A. Remond and C. Izard (eds.). Electrophysiological Effects of Nicotine. Amsterdam, Elsevier, 1979, pp. 133-146. (46) Fagerstrom, K.O., and Gotestam, K.G. Increase in muscle tonus after tobacco smoking. Addict. Behav. 2: 203-206, 1977. (47) Jones, R.T., Farrell, T.R., and Heming, R.I. Tobacco smoking and nice tine tolerance. In: Self-Administration of Abused Substances: Methods for Study (NIDA Research Monograph 20). Washington, D.C., U.S. Government Printing Office, 1978, pp. 202-208. (48) Henningfield, J.E. Pharmacologic basis and treatment of -cigarette smoking. J. Clin. Psychiatry 45: 24-34, 1984. (49) Austin, G.A. Perspectives on the History of Psychoactive Substance Use (NIDA Monograph 24). Washington, D.C., U.S. Government Print- ing Office, 1978. (50) Brecher, E.M. Licit and Illicit Drugs. The Consumers Union Report on Narcotics, Stimulants, Depressants, Inhalants, Hallucinogens, and Marijuana-Including Caffeine, Nicotine, and Alcohol. Boston, Little, Brown and Company, 1972. pp. 207-244. (51) Pomerleau, O.F., and Pomerleau, C.S. Neuroregulators and the rein- forcement of smoking Towards a biobehavioral explanation. Neurosci. Biobehav. Rev. 8: 503-513, 1984. (52) Wesnes, K., and Warburton, D.M. Smoking, nicotine and human per- formance. Pharmacol. Ther. 21: 189-234. 1982. (53) Wesnes, K., and Warburton, D.M. Smoking, nicotine and human per- formance. Pharmacol. Ther. 21: 189-208, 1983. (54) Wesnes, K.. and Warburton, D.M. The effects of cigarettes of varying yield on rapid information processing performance. Psychopharmacol- ogy,62:338-342,1984. (55) Williams, G.D. Effect of cigarette smoking on immediate memory and performance in different kinds of smokers. Br. J. Psychol. 71: 8390.1980. (56) Gilbert, R.M. Coffee, tea and cigarette use. Can. Med. Assoc. J. 120: 522-524, 1979. (57) Garvey, A. J., Bosse, R., and Seltzer, C.C. Smoking, weight change, and age. A longitudinal analysis. Arch. Environ. Health 28: 327-329.1974. (58) Heyden, S. The workingman's diet. Nutrition and Metabolism 20: 381-386, 1976. (59) Kittel, F., Rustin, R.M., Dramaix, M., DeBacker, G., and Komitzer, M. Psychosocial-biological correlates to moderate overweight in an indus- trial population. J. Psychosom. Res. 22: 145-158, 1978. (60) Jarvik, M.E. Nicotine and Appetite. Proceedings of the International Symposium on Tobacco Smoking and Health: A Neurobiological Approach (in press). (61) Glauser, S.C., Glauser, E .M., and Reidenberg, M.M. Metabolic changes associated with the cessation of cigarette smoking. Arch. Environ. Health 20: 377-381,197O. 171 (62) Schecter, M.D., and Cook, P.G. Nicotineinduced weight loss in rats without an effect on appetite. Eur. J. Pharmacol. 38: 63-69,1976. (65) Grunberg, N.E., and Morse, D.E. Cigarette smoking and food con- sumption in the United States. J. Appl. Psychol. (in press). (64) Burse. R.L.. Bynum, G.D., and Pandolf, K.B. Increased appetite and unchanged metabolism upon cessation of smoking with diet held con- stant. Physiologist 18: 157, 1975. (fT.5) Cherek, D.R. Effects of cigarette smoking on human aggressive behavior. Prog. Clin. Biol. Res. 169: 333-344, 1984. (66) Hughes, J.R., Hatsukami, D.K., Pickens, R.W., Krabn, D., Maline, S., and Luknic, A. Effect of nicotine on the tobacco withdrawal syndrome. Psychopharmacology 83: 82-87, 1984. (67) Grabowski, J., Stitzer, M.L., and Henningfield, J.E. Behavioral inter- vention techniques in drug abuse treatment (NIDA Research Mono- graph 46). Washington, D.C., U.S. Government Printing Office, 1984. (Ss) Blumberg, H.H.. Cohen, S.D., Dronfield, B.E., Mordecai, E.A., Roberts, J.C., and Hawks, D. British opiate users: I. People approach- ing London drug treatment centers. Int. J. Addict. 9: l-23, 1974. (69) Taylor, I.J., and `Ibylor, B.T. (eds.). Double Diagnosis: Double Dilemma. The Poly Addictions: Alcoholism, Substance Abuse, Smoking, and Gambling. J. Clin. Psychiatry (Suppl.) 45: l-44,1984. (70) Russell, M.A.H., Raw, M., and Jan&, M.J. Clinical use of nicotine chewing gum. Br. Med. J. 280: 1599-1602,198O. (71) Henning-field. J.E., Miyasato, K., and Jasinski, D.R. Abuse liability and pharmacodynamic characteristics of intravenous and inhaled nice tine. J. Pharmacol. Exp. Ther. 234: 1-12, 1985. (72) Henning-field, J.E., Miyasato, K., and Jasinski, D.R. Cigarette smokers self-administer intravenous nicotine. Pharmacol. Biochem. Behav. 19: 887-890, 1983. (73) Henning-field, J.E., and Goldberg, S.R. Control of behavior by in- travenous nicotine injections in human subjects. Pharmacol. B&hem. Behav. 19: 1021-1026, 1983. (74) Henningfield, J.E., and Goldberg, S.R. Nicotine as a reinforcer in human subjects and laboratory animals. Pharmacol. Biochem. Behav. 19:989-992, 1983. (75) Henning-field, J.E., and Goldberg, S.R. Stimulus properties of nicotine in animals and human volunteers: A review. In: L.S. Seiden and R.L. Balster (eds.). Behavioral Pharmacology: The Current Status. New York, Allan R. Liss, Inc., 1985, pp. 433-449. (76) Polin, W. The role of the addictive process as a key step in causation of all tobacco-related diseases. JAMA 252: 2874,1984. (77) U.S. Department of Health and Human Services, Public Health Ser- vice. Why People Smoke Cigarettes (PHS Publication No. 83-50195). Washington, D.C., U.S. Government Printing Office, 1983. 172 (78) U.S. Department of Health and Human Services. Drug Abuse and Drug Abuse Research. The First in a Series of `Biennial Reports to Con- gress (DHHS Publication No. ADM 851372). Washington, D.C., U.S. Government Printing Office, 1984, pp. 85-104. (79) Jasinski, D.R. Assessment of the abuse potentiality of morphine-like drugs (methods used in man). In: W.R. Martin (ed.). Handbook of Experimental Pharmacology, Vol. 45. Drug Addiction I. Berlin, West Germany, Springer-Verlag, 1977, pp. 197-258. (80) Martin, W.R. (ed.). Handbook of Experimental Pharmacology, Vol. 45. Drug Addiction I. Berlin, West Germany, Springer-Verlag, 1977, pp. 75-126. (81) American Psychiatric Association. Diagnostic and Statistical Manual of Mental Disorders (DSM-III). Washington, D.C., American Psychi- atric Association, 1980, p. 176. k?ZJ Shiffman, S.M. The tobacco withdrawal syndrome. In: N.A. Krasnegor led.). Cigarette Smoking as a Dependence Process (NIDA Research Monograph 23). Washington, D.C., U.S. Government Printing Office, 1979, pp. 158-184. (83) Gilbert, R.M.. and Pope, M.A. Early effects of quitting smoking. Psychopharmacology 78: 121-127, 1982. (84) Knapp, P.H., Bliss, C.M., and Wells, H. Addictive aspects in heavy cigarette smoking. Am. J. Psychiatry 119 966-972. 1963. (85) Ulett, J.A.. and Itil, T.M. Quantitative electroencephalogram in smok- ing and smoking deprivation. Science 164: 969-970.1969. (86) Knott, V.J., and Venables, P.H. EEG alpha correlates of nonsmokers, smoking, and smoking deprivation. Psychophysiology 14: 150-156, 1977. (87) Myrsten. A.L., Elgerot, A., and Edgren B. Effects of abstinence from tobacco smoking on physiological and psychological arousal levels in habitual smokers. Psychosom. Med. 39: 25-38, 1977. (88) Kleinman, K.M., Vaughn, R.L., and Christ, T.S. Effects of cigarette smoking and smoking deprivation on paired-associate learning of high and low meaningful nonsense syllables. Psychol. Rep. 32: 963-966, 1973. 039) Peterson, D.J., Lonegran, L.H., Hardinge, M.G., and I&l, C.W. Results of a stop-smoking program. Arch. Environ. Health 16: 211-214, 1968. (WJ Webrew, B.B., and Stark, J.D. Psychological and Physiological Changes Associated with Deprivation from Smoking. U.S. Naval Sub- marine and Medical Center Report No. 490, Bureau of Medicine and Surgery, Navy Department, 1967, pp. 1-19. (91) Shiffman, SM., and Jarvik, M.E. Smoking withdrawal symptoms in two weeks of abstinence. Psychopharmacology 50: 35-39, 1976. (92) Grabowski, J., and O'Brien, C.P. Conditioning factors in drug dependence: An overview. In: N. Mello (ed.). Advances in Substance Abuse: Behavioral and Biological Research, Vol. 2. Greenwich, Connecticut, JAI Press, 1981, pp. 69-121. 173 (93) Shiffman, SM., and Jarvik, M.E. Withdrawal symptoms: First week is the hardest. World Smoking Health 5: 15-21, 1980. (94) Jasinski, D.R. Opiate withdrawal syndrome: Acute and protracted aspects. AM. N.Y. Acad. Sci. 362: 183-186, 1981. (95) Hatsukami. D.K., Hughes, J.R., Pickens, R.W., and Svikis, D. Tobacco withdrawal symptoms: An experimental analysis. Psychopharmacol- ogy 84: 231-236, 1984. (%J Hughes, J.R., and Hatsukami, D. Signs and symptoms of tobaccowith- drawal. Arch. Gen. Psychiatry (in press). (97) Hatsukami, D.K., Hughes, J.R., and Pickens, R.W. Characteristics of tobacco abstinence: Physiological and subjective effects. In: J. Grabowski and S.M. HaIi teds.). Pharmacological Adjuncts in Smok- ing Cessation (NIDA Research Monograph 53). Washington, D.C., U.S. Government Printing Office, 1985. (%) Svikis, D.S., Hatsukami, D.K., Hughes, J.R., CarroII, K.M., and Pickens, R.W. Sex differences in tobacco withdrawal syndrome. Addict. Behav. (in press). (99) Hatsukami, D.K., Hughes, J.R., and Pickens, R.W. Blood nicotine, smoking exposure and tobacco withdrawal syndrome. Addict. Behav. (in press). (loo) Hoffmann, D., Harley, N.H., Fisenne, I., Adams, J.D., and Brunnemann, K.D. Carcinogenic agents in snuff. JNCI 76: 435-437,1986. (1OlJ Hoffmann. D., Hecht, S.S., Omaf, R.M., Wynder, E.L., and Iso, T.C. Chemical studies on tobacco smoke, XLII. Nitrosonornicotine: Presence in tobacco, formation and carcinogenicity. In: E.A. Walker, P. Bogovski, and L. Griciute (eds.). Environmental N-Nitroso Compounds. Analysis and Formation (IARC Scientific Publications No. 14). Lyon, France, International Agency for Research on Cancer, 1976, pp. 307-320. (102) Gritz, E.R., Baier-Weiss, V., Benowitz, N.L., Van Vunakis, H., and Jarvik, M.E. Plasma nicotine and cotinine concentrations in habitual smokeless tobacco users. CIin. Pharmacol. Ther. 30: 201-209, 1981. (103) Russell, M.A.H., Jarvis, M.J., Devitt, G., and Feyerabend, C. Nicotine intake by snuff users. Br. Med. J. 283: 814-817. 1981. (104) RusseII, M.A.H., Jarvis, M., West, R.J., and Feyerabend, C. BuccaIabsorp tion of nicotine from smokeless tobacco sachets. Lancet 8468: 1370,1985. (105) Hughes, J.R., Pickens, R.W., Spring, W.. and Keenan, R.M. Instruc- tions control whether nicotine wiIl serve as a reinforcer. J. Pharmacol. Exp. Ther. 235: 106-112, 1986. (106) Hatsukami, D.K., Gust, S.W., and Keenan, R. Physiological and subjec- tive changes from smokeless tobacco withdrawal. Manuscript submit- ted to JAMA, 1986. (107) Hughes, J.R., Hatsukami, D., and Skoog, K.P. Physical dependence on nicotine gum: A placebo substitution trial Paper presented at the Committee on Problems of Drug Dependence Meeting, Baltimore, Maryland, 1984. (108) West, R.J., and Russell, M.A. Effects of withdrawal from long-term nicotine gum use. Psychol. Med. 15: 891-893, 1985. 174 PHYSIOLOGIC AND PATHOGENIC EFFECTS OF NICOTINE AND SMOKELESS TOBACCO The user of smokeless tobacco is systematically exposed to signifi- cant amounts of nicotine, a potent multisystem pharmacologic agent. This chapter addresses the physiologic effects of nicotine upon the car- diovascular, nervous, and endocrine systems and the possible roles of nicotine in the pathogenesis of a variety of diseases. Nicotine is described in pharmacology textbooks as a stimulant of autonomic ganglia and skeletal neuromuscular junctions (i.e., nicotinic muscarinic receptors). However, in vivo the actions of nicotine are far more complex depending on the dose, target organ, prevalent auto nomic tone, and previous exposure history (tolerance) (1,Z). For pur- poses of this review, the focus is on the effects of nicotine in humans. Where human data are lacking and animal studies provide important information about physiologic effects, those studies are also discussed. Most data on the actions of nicotine in humans derive from studies of the effects of cigarette smoking, comparing cigarettes with and without nicotine, and studies of the effects of intravenous nicotine. These studies provide the basis for our understanding of the human pharma- cology of nicotine. However, as noted previously, actions of nicotine from smokeless tobacco and nicotine via inhalation or intravenous infu- sion may differ. Physiologic Effects of Nicotine Cardiovascular System The predominant cardiovascular actions of nicotine result from ac- tivation of the sympathetic nervous system. Smoking a cigarette in- creases the heart rate (10 to 20 BPM), blood pressure (5 to 10 mmHg), cardiac stroke volume and output, and coronary blood flow (34). Smok- ing may have different effects in smokers with coronary heart disease. It may reduce left ventricular contractility and cardiac output (6), ef- fects that are believed to be related to myocardial ischemia due to smoking-mediated tachycardia and the effects of carbon monoxide. Coronary blood flow may also decrease after smoking, which possibly is related to a nicotinemediated increase in coronary vascular resistance (7,8). Smoking, or nicotine intake, causes cutaneous vasoconstriction that is associated with a decrease in skin temperature, systemic veno- constriction, and increased muscle blood flow (411). Smoking results in increased circulating concentrations of norepi- nephrine, consistent with neural adrenergic stimulation, and epinephrine, indicating adrenal medullary stimulation (3). Circulating free fatty acids, glycerol, and lactate concentrations increase. Cardiovascular and metabolic effects are prevented by combined alpha and beta adrenergic blockade, which indicates that the cardiovascular effects of cigarette smoking are mediated by activation of the sympathetic nervous 175 system. Smoking-induced reduction in skin blood flow also can be antagonized by a vascular vasopressin antagonist, which suggests a role for vasopressin in mediating some cardiovascular responses (2). The cardiovascular effects of oral snuff have been examined system- atically in only one study (13). Changes in heart rate and blood pressure that are similar in magnitude to those of cigarette smoking were ob served. However, the time course appears to be slower than the response to cigarette smoking, with maximum effects observed at 5 to 10 minutes after a dose of oral tobacco. Similar findings, along with in- creased myocardial contractility and coronary, femoral, and renal blood flow, were also noted in anesthetized dogs after the administration of oral tobacco (13). Thus it appears that single doses of smokeless tobac- co can produce hemodynamic effects that are similar to those of cigarette smoking. Whether such changes are sustained throughout the day with repeated daily doses remains to be established. Central Nervous System Although smokers give different explanations for why they smoke, most agree that smoking produces arousal, particularly with the first few cigarettes of the day, as well as relaxation, especially in stressful situations (14). Desynchronization, decreased alpha and theta activity, and increased alpha frequency that is consistent with arousal are the usual electroencephalographic responses to cigarette smoking (X,16). These effects are blocked by mecamylamine, a centrally active nicotinic receptor antagonist, which indicates a role for nicotinic cholinergic receptor activation I1 7). Tobacco abstinence is associated with effects that are opposite those of smoking, namely, increased alpha power and reduced alpha frequency (15,18). Endocrine System Cigarette smoking and nicotine have been reported to increase circu- lating levels of catecholamines, vasopressin, growth hormone, cortisol, ACTH, and endorphins (3,19,20). Nicotine inhibits the synthesis of prostacyclin in rabbit aorta and human peripheral veins and the hypoxia-induced release of prostacyclin from rabbit hearts (21). Cigarette smoking has been reported to decrease the urinary excretion of prostacyclin met&o&s in humans, which sup ports the prediction from animal studies 122). F'rostacyclin has anti- aggregatory and vasodilating actions that are believed to play a homeo static role in preventing vascular thrombosis. Nicotine, Smokeless Tobacco, and Human Diseases As attested to in the Surgeon General's reports since 1964, smoking is a major risk factor for coronary and peripheral vascular disease, cancer, chronic obstructive lung disease, peptic ulcer disease, and repro 176 ductive disturbances, including prematurity. Tobacco smoke is a com- plex mixture of chemicals, including carbon monoxide, many of which are believed to contribute to human disease. Smokeless tobacco likewise exposes users to a number of chemicals, particularly nicotine. Nicotine may play a contributory or supportive role in the pathogenesis of many smoking-related diseases. That nicotine causes human disease de novo has not been proven; however, its potential health conse quences deserve serious consideration. More direct data on its causal role are needed. Coronary and Peripheral Vascular Disease Nicotine may contribute to atherosclerotic disease by actions on lipid metabolism, coagulation, and hemodynamic effects. Compared to non- smokers, cigarette smokers have elevated levels of low density (LDL) and very low density lipoproteins (VLDLI and reduced levels of high density lipoproteins (HDL). This profile is associated with an increased risk of atherosclerosis (23). It is hypothesized that nicotine, by releasing free fatty acids, increases the synthesis of triglycerides and VLDL by the liver, which in turn results in decreased HDL production. In most studies, the blood of smokers is shown to coagulate more easily (24), platelets are found to be more reactive, and platelet survival is shortened when compared to nonsmokers (25,). Thrombosis is believed to play a role that promotes the growth of vascular endothelial cells that contribute to the atherosclerotic plaque. The importance of nicotine as a determinant of platelet hyperreactivity is supported by a study that shows an apparent relationship between nicotine concentrations after smoking different brands of cigarettes and platelet aggregation response (26). Nicotine may affect platelets by releasing epinephrine, which is known to enhance platelet reactivity; by inhibiting prosta- cyclin, an antiaggregatory hormone that is secreted by endothelial cells; or perhaps directly. Finally, by increasing the heart rate and cardiac output, nicotine increases blood turbulence and may promote endo thelial injury. Although several potential mechanisms for promoting atherogenais have been considered, nicotine has not yet been demon- strated to accelerate atherosclerosis in experimental animals. Nicotine may play a role in causing acute coronary events. Myocar- dial infarction can occur with one or more of three precipitants: throm- bosis, excessive oxygen and substrate demand, and coronary spasm. Nicotine can promote thrombosis as discussed previously. Nicotine in- creases the heart rate and blood pressure and, therefore, myocardial ox- ygen consumption Coronary blood flow increases in a healthy person to meet the increased demand. In the presence of coronary heart disease, ischemia may develop and myocardial dysfunction may occur. Nicotine may induce coronary spasm by sympathomirnetic actions or by the inhibition of prostacyclin. Coronary spasm has recently been reported to occur during cigarette smoking (27). All of the above may contribute 177 to the precipitation of acute myocardial infarction in a person with pre existing coronary atherosclerosis. Cigarette smoke exposure decreases the ventricular fibrillation threshold after experimental myocardial infarction in dogs (28). How much of this effect is due to nicotine and how much is due to carbon monoxide have not been established. Sudden cardiac death in smokers might result from ischemia, as discussed above, combined with the arrhythmogenic effect of increased circulating catecholamines. Cigarette smoking has not been associated with an increased prevalence of hypertension. However, a recent preliminary report sug- gested higher blood pressure in young men who used smokeless tobacco compared to cigarette smokers or nonsmokers (25% Smokers who have essential hypertension experience an accelerated progression of vascular and renal disease. Nicotine may contribute to such a process by producing vasoconstriction or enhancing coagulation There also may be other interactions with hypertensive disease. For example, a pa- tient with a pheochromocytoma developed paroxysmal hypertension and angina pectoris following the use of oral snuff (Z@). In a controlled situation, blood pressure was recorded to increase from 110/70 to 300/103 with a heart rate increase from 70 to 110 within 10 minutes after the use of oral snuff. Rechallenge after surgery for the pheochro mocytoma revealed only the usual blood pressure increase. Peptic Ulcer Disease Smoking is strongly related to the prevalence of peptic ulcer disease, and failnre to stop smoking is the major predictor of failure to respond to ulcer therapy (31). Smoking decreases pancreatic fluid and bicar- bonate secretion that result in greater and more prolonged acidity of gastric fluid of the duodenal bulb (32). Similar effects after the infusion of nicotine have been reported in animals (33). The swallowing of tobac- co juice that contains large concentrations of nicotine may conceivably have local effects and therefore elicit added concern for the use of smokeless tobacco. Smoking is a major risk factor for low birth weight and, conse quently, fetal morbidity and mortality (34). `Ibbacco smoke may influ- ence the fetus either through alterations in maternal physiology that limit the nutrient flow to the fetus or by the transplacental passage of smoke components that have direct effects on the fetus. The factors that are considered most likely to affect the fetus are carbon monoxide and nicotine. Carbon monoxide inhalation has been shown to increase carboxyhemoglobin in both maternal and fetal blood that possibly limits oxygen supply to the fetus (35) However, while newborn infants 178 of smoking mothers have higher concentrations of carboxyhemo globin than do neonates of nonsmokers, there are only trivial differ- ences in hemoglobin concentrations, hematocrit, and various charac- teristics of hemoglobin (36). Thus it is difficult to explain an adverse effect that is based on chronic hypoxia due to carbon monoxide in tobacco smoke. It is more likely that nicotine is important in causing adverse effects. The effects of nicotine on the fetus may include a reduction of uterine blood flow or a direct effect on fetal function (3738). The presence of nicotine and its principal metabolites has been demonstrated in the um- bilical cord blood and urine of newborn infants of smoking mothers, as well as in amniotic fluid, indicating transplacental passage (39). Nonnicotine-Related Adverse Metabolic Consequences certain brands of chewing tobacco and snuff contain glycyrrhizinic acid, which is also an ingredient of licorice. Glycyrrhizinic acid has po tent mineralocorticoid hormone activity that can result in potassium wasting. `l%ro patients who were heavy users of oral smokeless tobacco developed severe hypokalemia with muscle weakness (and in one case, evidence of muscle breakdown) that apparently was due to the ingestion of large amounts of this substance (40). Smokeless tobacco also contains large amounts of sodium (41) that, if swallowed, may aggravate hyper- tension or cardiac failure. References (1) Cornroe, J.H. The pharmacological actions of nicotine. Ann. N.Y. Acad. sci. 90: 48-51, 1960. (2) Su, C. Actions of nicotine and smoking on circulation. Pharmacol. Ther. 17.. 129141,1982. (3) Cryer, P.E., Haymond, M.W., Santiago, J.V., and Shah, S.D. Norepi- nephrine and epinephrine release and adrenergic mediation of smoking- associated hemodynamic and metabolic events. N. Engl. J. Med. 2%: 573-577, 1976. (4) Irving, D.W., and Yamam oto, T. Cigarette smoking and cardiac out- put. Br. Heart J. 25 126-132, 1963. (5) Bargeron, L.M., Ehmke, D., Gonlubol, F., Castellanos, A., Siegel, A., and Bing, R.J. Effect of cigarette smoking on coronary blood flow and myocardkl metabolism. Circulation 15: 251-257,1957. (6) Pentecost, B., and Shillingford, J. The acute effects of smoking on myocardial performance in patients with coronary arterial disease. Br. Heart J. 26: 422-429, 1964. (7) Klein, L.W., Ambrose, J., Pichard, A., Holt, J., Gorlin, R., and Teichholz, L.E. Acute effects of cigarette smoking on coronary vas- cular dynamics. Circulation (Abst.) 68: 165, 1983. 179 (8) Reddy. C.V.R.. Khan, R.G., Feit, A., Chowdry, I.H., and El Sherif, N. Effects of cigarette smoking on coronary hemodynamics in coronary artery disease. Circulation (Abst.) 68: 165, 1983. (9) Freund, J., and Ward, C. The acute effect of cigarette smoking on the digital circulation in health and disease. Ann. N.Y. Acad. Sci. 90: 85-101, 1960. (10) Eckstein, J.W., and Horseley, A.W. Responses of the peripheral veins in man to the intravenous administration of nicotine. Ann. N.Y. Acad. Sci. %l: 133-137, 1960. (11) Rottenstein, H., Pence, G., Russ, E.. Felder, D., and Montgomery, H. Influence of nicotine on the blood flow of resting skeletal muscle and of the digits in normal subjects. Ann. N.Y. Acad. Sci. %? 102-113, 1960. (12) Waeber, G., Schaller, M., Nussberger, J., Bussien, J., Hofbauer, K.G., and Brunner, H.R. Skin blood flow reduction induced by cigarette smoking Role of vasopressin. Am. J. Physiol. 247: H895-H901, 1984. (13) Squires, W.G., Branton, T.A., Zinlcgraf, S., Bonds, D., Hartung, G.H., Murray, T., Jackson, A.S., and Miller, R.R. Hemodynamic effects of oral smokeless tobacco in dogs and young adults. Prev. Med. 13: 195-206, 1984. (14) Henningfield, J.E. Behavioral pharmacology of cigarette smoking. In: T. Thompson, T.B. Dews, and J.E. Barrett (eds.). Advances in Behav- ioral Pharmacology, Vol. IV. New York, Academic Press, 1984, pp. 131-210. (15) Heming, R.I., Jones, R.T., and Bachman, J. EEG changes during tobacco withdrawal. Psychophysiology 20: 507-512, 1983. (16) Knott, V.J., and Venables, P.H. EEG alpha correlates of nonsmokers, smokers and smoking deprivation. Psychopharmacology 14: 150-156, 1977. (17) Domino, E.F. Behavioral electrophysiological, endocrine, and skeletal muscle actions of nicotine and tobacco smoking. In: A. Remond and C. Izard (eds.). Electrophysiological Effects of Nicotine. Amsterdam, ElsevieriNorth Holland Biomedical Press, 1979, pp. 133-146. (18) Ulett, J., and Itil, T. Quantitative electroencephalogram in smoking and smoking deprivation. Science lf%: 969-970,1969. (19) Sandberg, H., Roman, L., Zavodnick, J., and Kupers, N. The effect of smoking on serum somatotropin, immunoreactive insulin and blood glucose levels of young adult males. J. Pharmacol. Exp. Ther. 184: 787-791, 1973. (20) Pomerleau, O.F., Fertig, J.B., Seyler, L.E., and Jaffe, J. Neuroendo crine reactivity to nicotine in smokers. Psychopharmacology 81: 61-67, 1983. (21) Wennmalm, A. Nicotine inhibits hypoxia- and arachidonateinduced release of prostacyclin-like activity in rabbit hearts. Br. J. Pharmacol. 69 545-549, 1980. (22) Nadler, J.L., Velasco, J.S., and Horton, R. Cigarette smoking inhibits prostacyclin formation. Lancet 1: 1248-1250, 1983. (23) Brischetto, C.S., Connor, W.E., Connor, S.L., and Matarazzo, J.D. Plasma lipid and lipoprotein profiles of cigarette smokers from ran- domly selected families: Enhancement of hyperlipidemia and depres- sion of high-density lipoprotein. Am. J. Cardiol. 52: 675-680, 1983. (24) Bilhmoria, J.D., Pozner, H., Metselaar, B., Best, F.W., and James, D.C.O. Effect of cigarette smoking on lipids, lipoproteins, blood coagulation, fibrinolysis and cellular components of human blood. Atherosclerosis 21: 61-76, 1975. (25) Mustard, J.F., and Murphy, E.A. Effect of smoking on blood coagula- tion and platelet survival in man. Br. Med. J. 1: 846-849, 1963. (26) Renaud, S., Blache, D., Dumont, E., Thevenon, C., and Wissendanger, T. Platelet function after cigarette smoking in relation to nicotine and carbon monoxide. Clin. Pharmacol. Ther. 36: 389-395, 1984. (27) Maouad, J., Fernandez, F., Barrillon, A., Gerbaux, A., and Gay, J. Dif- fuse or segmental narrowing (spasm) of the coronary arteries during smoking demonstrated on angiography. Am. J. Cardiol. 53 354-355, 1984. (`28) Bellet, S., DeGuzman, N.T., Kostis, J.B., Roman, L., and Fleischmann, D. The effect of inhalation of cigarette smoke on ventricular fibrillation threshold in normal dogs and dogs with acute myocardial infarction. Am. Heart J. 83: 67-76, 1976. (29) Schroeder, K.L., and Chen, M.S. Smokeless tobacco and blood pres- sure. N. Engl. J. Med. 312: 919, 1985. (30) McPhaul, M., Punzi, H.A., Sandy, A., Borganelli, M., Rude, R., and Kaplan, N.M. Snuff-induced hypertension in pheochromocytoma. JAMA 252: 2860-2862.1984. (311 Korman, M.G., Shaw, R.G., Hansky, J., Schmidt, G.T., and Stem, A.I. Influence of smoking on healing rate of duodenal ulcer in response to cimetidine or high-dose antacid. Gastroenterology 80: 1451-1453,198l. (32) Mm-thy, S.N.S., Dinoso, V.P., Clearfield, H.R., and Chey, W.Y. Simultane ous measurement of basal pancreatic, gastric acid secretion, plasma gastrin, and secretin during smoking. Gastroenterology 73: 758-761, 1977. (33) Konturek, S.J., Dale, J., Jacobson, E.D., and Johnson, L.R. Mecha- nisms of nicotine-induced inhibition of pancreatic secretion of bicar- bonate in the dog. Gastroenterology 62: 425-429, 1972. (34) Abel, E.L. Smoking during pregnancy: A review of effects on growth and development of offspring. Hum. Biol. 52: 593-625, 1980. (35) Longo, L.D. The biological effects of carbon monoxide on the pregnant woman, fetus and newborn infant. Am. J. Obstet. Gynecol. l.29: 69,1977. (36) Bureau, M.A., Shapcott, D., Berthiaume, Y., Monette, J., Blovin, D., Blanchard, P., and Begin, R. A study of P50,2,3diphosphoglycerate, total hemoglobin, hematocrit and type F hemoglobin in fetal blood. Pediatrics 72: 22, 1984. 181 (37) Ayromlooi, J., Desiderio, D., and Tobias, M. Effect of nicotine sulfate on the hemodynamics and acid base balance of cbronicaIIy instru- mented pregnant sheep. Dev. Pharmacol. Ther. 3: 205-213, 1981. (38) Resnik, R. Brink, G.W., and Wilkes, M. Catecholaminemediatedreduc- tion in uterine blood flow after nicotine infusion in the pregnant ewe. J. Clin. Invest. 63: 1133-1136, 1979. (39) Hibberd, A.R., O'Connor, V., and Gorrod, J.W. Detection of nicotine, nicotine-l `-N-oxide and cotinine in maternal and fetal body fluids. In: J.W. Gorrod (ed.). Biological Oxidation of Nitrogen. Amsterdam, Elsevier, 1978, pp. 353-361. 140) Valeriano, J., Tucker, P., and Kattab, J. An unusual cause of hypoka- lemic muscle weakness. Neurology 33: 1242-1243, 1983. (41) Hampson, N.B. Smokeless is not saltless. N. Engl. J. Med. 31.2: 919, 1985. CONCLUSIONS 1. The use of smokeless tobacco products can lead to nicotine dependence or addiction. 2. An examination of the pharmacokinetics of nicotine (i.e., nico- tine absorption, distribution, and elimination) resulting from smoking and smokeless tobacco use indicates that the magni- tude of nicotine exposure is similar for both. 3. Despite the complexities of tobacco smoke self-administration, systematic analysis has confirmed that the resulting addiction is similar to that produced and maintained by other addictive drugs in both humans and animals. Animals can learn to dis- criminate nicotine from other substances because of its effects on the central nervous system. These effects are related to the dose and rate of administration, as is also the case with other drugs of abuse. 4. It has been shown that nicotine functions as a reinforcer under a variety of conditions. It has been confirmed that nicotine can function in all of the capacities that characterize a drug with a liability to widespread abuse. Additionally, as is the case with most other drugs of abuse, nicotine produces effects in the user that are considered desirable to the user. These effects are caused by the nicotine and not simply by the vehicle of delivery (tobacco or tobacco smoke). 5. Nicotine is similar on ah critical measures to prototypic drugs of abuse such as morphine and cocaine. The methods and criteria used to establish these similarities are identical to those used for other drugs suspected of having the potential to produce abuse and physiologic dependence. Specifically, nicotine is psycho- 182 active, producing transient doserelated changes in mood and feeling. It is a euphoriant that produces doserelated increases in scores on standard measures of euphoria. It is a reinforcer lor reward) in both human and animal intravenous self-administration paradigms, functioning as do other drugs of abuse. Additionally, nicotine through smoking produces the same effects, and it causes neuroadaptation leading to tolerance and physiologic dependence. l&en together, these results confirm the hypothesis that the role of nicotine in the compulsive use of tobacco is the same as the role of morphine in the compulsive use of opium derivatives or of cocaine in the compulsive use of coca deriva- tives. 6. The evidence that smokeless tobacco is addicting includes the pharmacologic role of nicotine dose in regulating tobacco intake; the commonalities between nicotine and other prototypic dependenceproducing substances; the abuse liability and depen- dence potential of nicotine; and the direct, albeit limited at pres- ent, evidence that orally delivered nicotine retains the character- istics of an addictive drug. 7. Several other characteristics of tobacco products in general, including smokeless tobacco, may function to enhance further the number of persons who are afflicted by nicotine dependence: nicotine-delivering products are widely available and relatively inexpensive; and the self-administration of such products is legal, relatively well tolerated by society, and produces minimal disrup- tion to cognitive and behavioral performance. Nicotine produces a variety of individual-specific therapeutic actions such as mood and performance enhancement; and the brief effects of nicotine ensure that conditioning occurs, because the behavior is associated with numerous concomitant environmental stimuli. 8. All commonly marketed and consumed smokeless tobacco prod- ucts contain substantial quantities of nicotine; the nicotine is delivered to the central nervous system in addicting quantities when used in the fashion that each form is commonly used (or as recommended in smokeless tobacco marketing campaigns). 9. Since the exposure to nicotine from smokeless tobacco is similar in magnitude to nicotine exposure from cigarette smoking, the health consequences of smoking that are caused by nicotine also would be expected to be hazards of smokeless tobacco use. Areas of particular concern in which nicotine may play a contributory or supportive role in the pathogenesis of disease include coronary artery and peripheral vascular disease, hypertension, peptic ulcer disease, and fetal mortality and morbidity. 183 Available data clearly support the view that nicotine produces behav- ioral and physiologic dependence and has effects on all critical dimen- sions exemplified by a drug with a profile of high abuse liability. Never- theless, the resolution of several questions is essential. These questions revolve around the relationships between the several forms of tobacco use. They parallel and have commonalities with important issues in other forms of drug abuse (e.g., cocaine). There are several major msearch areas that could provide data of potential public health significance. The first area of research is the relationship between the rate of nice tine administration and abuse liability. Existing data suggest that the slowest commercially available nicotinereleasing preparation, nicotine gum, has a lower abuse liability than the fastest commercially available nicotine-releasing ppparation, cigarettes. These facts further suggest the possibility that there might be quantifiable differences in abuse liability among tobacco product forms. The second area of research importance involves the relationship be- tween the initiation of one form of tobacco use, e.g., smokeless tobacco, and the use of other forms of tobacco, e.g., cigarettes. The relationships between common forms of tobacco use, the extent to which they are interchangeable, and the possibility that the use of one form of tobacco leads to the use of another need examination. A third area of specific importance relates to the extent to which tobacco use, with its implicit acceptance, encourages other drug use. A related question is the extent to which exposure to drug effects, both neurologic and behavioral, modifies subsequent drug responses or establishes the conditions for other equally harmful drugs to become reinforcers. These issues follow from the observations that cigarette use is a major correlate (possibly a "stepping stone") of other kinds of drug dependence and that regular tobacco use generally leads to other forms of drug addiction. A fourth area of research is prevention and treatment. Recent sur- veys indicate that youth attribute negligible risk to smokeless tobacco products, suggesting the possible need for education-based prevention approaches. Regarding treatment, it is plausible that nicotine gum treatment could be of even greater relative utility for smokeless tobacco users than for cigarette smokers because of the more similar pharmaco kinetic profiles of smokeless tobacco and gumdelivered nicotine com- pared to cigarette smoke-delivered nicotine. The absorption and distribution characteristics of nicotine with the use of smokeless tobacco may differ from those of cigarette smoking. The pharmacodynamic and pharmacologic consequences of such dif- ferences may be important but require additional future research FLU- ther studies to define more precisely the role of nicotine and of smokeless tobacco in the causation of diseases other than those that 184 involve the oral cavity are clearly needed. Specifically, research is needed to: o Determine nicotine blood levels and time course in various popula- tions of smokeless tobacco users, including established users. o Determine the cardiovascular, hormonal, and metabolic effects of smokeless tobacco when used in a regular fashion throughout the day. o Determine the influence of the rate of absorption of nicotine on the effects from smoking cigarettes and the use of smokeless tobacco. o Using experimental studies, determine the effects of smokeless tobacco in users of different ages and high-risk status (i.e., patients with hypertension, coronary heart disease, peripheral vascular disease, and peptic ulcer). o Using epidemiologic studies, determine the risk potential of the regular use of smokeless tobacco on the development of diseases such as coronary heart disease, peptic ulcer, and complications of Pregnancy. 185 INDEX ABBREVIATIONS constituents of smokeless tobacco, 70 ABRASION (DENTAL) defined, 128 relation to smokeless tobacco, 130 ABUSE LIABILITY OF NICOTINE defined, 145,157 evidence for abuse potential, 166-167 experimental studies, 157-166 implications of studies, 162 phannacodynamic effects, 158 research needs, 184 self-administration, 159-l 62 ACANTHOSIS clinical report, 115-118 defined, 113 ADENOCARCINOMA relation to snuff use, 48 ADOLESCENT USE attitudes, 24 data criteria, 17 ethnic groups, 20 males vs. females, 20 National Program Inspection study, 24 peer and family influences, 24 rural vs. urban areas, 20 ADULT USE patterns, 16-17 ADVISORY COMMIlTEE TO THE SURGEON GENERAL, REPORT carcinogens revealed, vii conclusions, vii-viii, xxii gingival recession, viii historical perspective of report, Xvii-xix nicotine addiction and effects, viii noncancerous conditions, xxi oral leukoplakia. viii organization of report, xvii overview, xx-xxii precancerous oral health conditions, xxi review methods, xix-xx ALCOHOL CONSUMPTION relation to smokeless tobacco, 35 ANIMAL MODELS bioassays with chewing tobacco, 79, 82-83 bioassays with constituents of smokeless tobacco, 86-88 bioassays with snuff, 83-86 metabolism of NMOR, 74-75 metabolism of NNK, 71-73 metabolism of NNN, 73-74 mutagenicity assays, 88-89 pharmacodynamic effects of nicotine, 158 physiologic effects of nicotine, 178 sdivmy glands, 126-127 short-term tests, 88-89 ASIAN DATA case-control studies, 42-43, 119 chewing of quids, 42-43, 50 esophageal neoplasms, 50 rates of oral cancer, 42-43 smokeless tobacco products, 42-43 smoking and oral cancer risk, 43-44 BIOASSAYS FOR CL4RCTVOGEVIC Ati'UK'TY OF CZEWiVG TOBACCO OR tZi%`W;wG iYm4ccoIiYxmAcr$8o BIOASSAYS FOR CARClhDGEVIC ACTNITY OF Sh?UFF OR SNUFF EXlTUCI$81 BIOASSAYS WITH CHEWING TOBACCO (See dso ANIMAL MODELS) application to the cheek pouch, 79, 82-83 application to the oral mucosa, 79, 82-83 oral administration, 79 skin application, 83 subcutaneous application, 83 BJOASSAYS WITH CONSTITUENTS OF SMOKELESS TOBACCO (See also ANIMAL MODELS) inducement of tumors, 86-88 187 BIOASSAYS WITH SNUFF (See also ANIMAL MODELS) application to the lip, oral mucosa, or cheek pouch, 84-86 oral administration, 83-84 relation to herpes simplex virus, 86-87 subcutaneous administration, 86 Bladder Neophsms, See URINARY TRACT NEOPLASMS Blockade Therapy, See NICOTINE ADDICTION TREATMENT BUCCAL CARCINOMA case studies, 34-36 CALCULUS (DENTAL) defined, 129 use of smokeless tobacco, 130 CARC~OGHCflY OF TOBACCO SPECIFIC NITROSAMINES 82 CARCINOGENS daily exposure from snuff and chewing tobacco usage, 64 in smokeless tobacco, 58-60. 64-65 N-nitrosamines, 58-60, 64 polonium-210 (21oPo), 58, 65 polynuclear aromatic hydra carbons (PAH), 58,65 saliva analysis, 64 CARCINOMA hVSlTU defined, 114 diagnoses differ, 123 squamous cell carcinoma study, 120 Cardiovascular System, See PHYSIOLOGIC EFFECTS OF NICOTINE Central Nervous System, See PHYSIOLOGIC EFFECTS OF NICOTINE ChXRACTERLSTICS OF SMOKE I2239 TOBACCO PRODUCT$6 CHEEK NEOPLASMS (See also ORAL CANCER) animal models, 79, 82, 84 case study, 120 CHEMICAL COMPOSITION processed tobacco, 58 nicotine, 58 CHEMICAL CONSTITUENTS OF SMOKELESS TOBACCO carcinogens, 58-59,60,6465 chemical composition, 58 CHEVRON KERATINIZATION case report, 116-l 17 defined, 114 CHEWING TOBACCO (See also SMOKELESS TOBACCO, BIOASSAYS) case-control studies, 36,40 characteristics, 5 looseleaf, 5,7 plug, 5, 7 twist, 5, 7 CommonaIities Between Tobacco and Other Addictive Substances, See NICOTINE ADDICTION CONCEPTUAL MODEL of smokeless tobacco lesions, 121 CONCEPTUAL NflVRAL HISTORY OF ORAL MucosAL CRANGES ASlSOCiWED WITH TdcE USE OF SMO- TOBACCO, 122 CORONARY AND PERIPHERAL VASCULAR DISEASE acute coronary events, 177-178 and nicotine, 177-178 atherosclerosis, 177 effect on platelets, 177 endothelial injury, 177 research needed, 185 ventricular fibrillation, 178 Cotinine, See PHARMACQ KINETICS OF NICOTINE CURRENT POPULATION SURVEY (CPS) survey data, 12,14 DATA: ABUSE LUBIUTY .!U'tLVm AT ADDICTION RESEARCH CENTER 160 DENTAL CARIES defined, 128 relation to smokeless tobacco, 129-130 DOSE RESPONSE EFFECTS (See also ANIMAL MODELS) and oral leukoplakia, 13 1 nicotine, 147, 152-153, 158, 182-183 DRUG DEPENDENCE commonalities between tobacco and other substances, 146-147, 152-157 defined, 145 physiologic or physical dependence, 145-146 188 DYSPLASIA defined, 114 diagnoses differ, 121. 123 relation to oral lesions, 115-l 16 Endocrine System, See PHYSIOLOGIC EFFECTS OF NICOTINE EPIDEMIOLOGIC STUDIES AND SMOKELESS TOBACCO data from Asia, 42-44 data from North America and Europe, 33-42 oral cancer, 33-44 other cancers, 47-55 EROSION (DENTAL) defined, 128 ERYTHROPLAKIA defined, 113 ESOPHAGEAL NEOPLASMS case-control study, 39, 48-50 cohort study, 49-50 ESTMTED E2XkX?RE OF US RESIDm To NITROSmES 64 ESTLlzATED RELATWE RISKS ASSOCL4TED WITH SNUFF USE FOR ClANCERS OF TljlE ORAL CAV7TI: PHARYNX, AND LARYNX, CASECONTROL DATA FROM VOGLER ETAL., Fm ONLY 38 EST~TEDRELAT~RLSKFOR CMCER OF THEHEADAND NECK FROM SMOKELESS TOBACCO USE BY ANATOMIC SITE, Tm NATIONAL CAN- CER SUR VEX MALES ONLY 39 EST~TEDRELAT~RLSK OF OROPHARYNGEU CANCER ACCORDING To DURATION OF SNUFF USEANDANAToM7C SITE, Wm ET AL, 41 ESTWTES OF RELATNE RISKS OF BLADDER CANC' LN PERSONS WHO HA VE EVER USED C%!EW~G TOBACCO AND SNUFF 53 ETHNIC USE OF SMOKELESS TOBACCO of youth, 21-22 Experimental Studies Exposing Laboratory Animals, See ANIMAL MODELS FORMATION OF TOBACCO SPECUIC NITROSmES 60 FREQUENCY OF SMOKELESS TOBACCO USE LN PAST YEARi 14 Gastric Neoplasms, See STOMACH NEOPLASMS GENERAL PRINCTPLESLN EVALUATmTG CARCXNOGENIC RISK OF CHEMC~S OR COM- PLEX MLX-TURES (INTERNA- TIONAL AGENCY FOR RESEARCH ON CANCER, xxi GINGIVAL AND PERIODONTAL HEALTH bone loss, 125 effects of smokeless tobacco, 101, 103-104, 123 inflammation, 126 studies in Sweden, 126 studies in the United States, 125 GINGIVAL CARCINOMA case studies, 34, 36, 44, 120 GINGIVAL RECESSION Advisory Committee report, viii defined, 123 related to oral leukoplakiai mucosal pathology, 112 studies in the United States, 125 GINGIVITIS defined, 123 relation to smokeless tobacco, 125, 129 studies in the United States, 125 GLYCYRRHIZIC ACID metabolic consequences, 179 HEAD AND NECK NEOPLASMS case-control study, 38-39.38-39 HEROIN ADDICTION THERAPY model used, 156 HERPES SIMPLEX VIRUS relation to oral cancer, 86-87, 93 HISTORICAL PERSPECTIVE chewing tobacco, xvii consequences of use, xviii-xix decline of use, 5 early uses, xviii-xix, 5 snuff, xvii-xix HYPERKERATOSIS ccrhort study, 117-l 18 defined, 113 relation to snuff use, 125 189 HYPERORTHOKERATOSIS cohort study, 118 defined, 113 HYPERPARAKERATOSIS cohort study, 118-l 19 defined, 113 HYPOKALEMIA nonnicotine metabolic consequences, 179 INHALED SNUFF relation to nasal carcinoma, 93 Kidney Neoplasms, See URINARY TRACT NEOPLASMS LARYNGEAL NEOPLASMS /See also THIRD NATIONAL CANCER STUDY) case-control study, 37,38, 50-51 Leukoplakia, Oral, See ORAL LEUKOPLAKIA LIP NEOPLASMS (See also ORAL CANCER) animal models, 84-85 case-control study, 36 labial mucosa, 116-117 Looseleaf, See CHEWING TOBACCO MANLT?ACTURLNG TRENDS QUANTLTLES OF SMORELESS TOBACCO MANUFACTURED LN THE UiWTED STATES FROM 1961 To 1985 EXPRESSED LN JDLLION POUNDS. 8 MEAN FREQUENCY OF SMOKE LESS TOBACCO USE DURING LAST 7 DAYS BY ETHNLCITY BY MALE RESPONDENTS, 23 METABOLIC PATHWAYS OF NMOR, 74 ME;X&Xl$IC PATHWAYS OF ,A . METkOLIC PATmA YS OF Nvy 73 METABOLISM OF CONSTITUENTS OF SMOKELESS TOBACCO conversion of products, xx-xxi of NMOR, 74-75 of NNK, 71-73 of NNN, 73-74 MOUTH NEOPLASMS (See also ORAL CANCER) case-control study, 36-37.36, 37, 39,120 MUCOSAL PATHOLOGY snuff-induced lesions, 114-l 16 studies in Scandanavia, 11 l-l 12 studies in the United States, 107, 108-109110-112 MUTAGENICITY ASSAYS AND OTHER SHORT-TERM TESTS (See also ANIMAL MODELS) chewing tobacco, 88 snuff, 88 tobacco-specific N-nitrosamines, 88-89 NASAL NEOPLASMS posterior nasal space tumors, 48 relation to snuff use, 48 Nass, See ASIAN DATA NATIONAL BLADDER CANCER STUDY relation to smokeless tobacco use, 52-53 NATIONAL BLOOD CANCER STUDY survey data, 16 NATIONAL HEALTH INTER- VIEW SURVEY (NHIS) survey data, 10, 14-15 NATIONAL INSTITUTE ON DRUG ABUSE (NIDA) survey data, 13-15 NATIONAL PREVALENCE OF CURRENT USE OF SNUFF BY GENDER AGE, AND RACE FOR 1980 THROUGH 1985,ll NATION& PREVALENCE OF SMOEELESS TOBACCO USE BY ADCZT STATUS AND SEX, INIDA SAMPLE, 1935, 13 NATIONAL PREVXLENCE OF SMOKES TOBACCO USE: DATA SOURCES, 9 NATIONAL SURVEY DATA conclusions, 24-25 Current Population Survey, 12,14 discussion of data, 14-15 National Health Interview survey, 10, 14-15 National Institute on Drug Abuse Household Survey, 13-15 Office on Smoking and Health Surveys, 7,10,12,14 Simmons Market Research Bureau, 10, 12, 14-15 190 NEUROADAPTATION (See also PHYSICAL DEPENDENCE POTENTIAL OF NICOTINE, PHYSIOLOGIC EFFECTS OF NICOTINE) defined, 145. demonstrated, 153 nicotine, 162 NICOTINE (See also PHARMA. COKINETICS OF NICOTINE, NICOTINE ADDICTION, NICOTINE EXPOSURE, ABUSE LIABILITY, PHAR- MACODYNAMICS OF NICO- TINE, PATHOLOGIC EFFECTS OF NICOTINE, AND SMOKELESS TOBACCO) absorption, 141-142, 185 cotinine levels, 143 dependence, 154 distribution, 142 elimination, 142-143 habituating agent, 58 levels, 143 physical dependence, 162-166 self-administration, 159-161 therapeutic effects, 154-155 turnover, 143 NICOTINE ADDICTION /See also NICOTINE EXPOSURE, PHYSICAL DEPENDENCE POTENTIAL OF NICOTINE) commonalities with other addictive substances, 146-147, 152-157, 184 dose manipulations, 153 encourages other drug use, 184 pattern of use, 152 relation to smokeless tobacco, 58, 144-147, 152, 185 self-administration, 152-153 social tolerance, 156 therapeutic effects, 154-155 tolerance of use developed, 153 treatment for, 155-156 NICOTINE ADDICTION TREATMENT blockade therapy, 156 nicotine gum, 156 pretreatment, 156 substitution, 156 supportive therapy, 156 NICOTINE CHEWING GUM as substitution, 156 reinforcing properties, 166-167 research needs, 184 self-administration, 167 NICOTINE EXPOSURE /See also NICOTINE, NICOTINE ADDICTION) delivery to the central nervous system, 147 other physiological effects, . vm, xxii, xxvi pharmacokinetics, xxiv, 141-144 peer pressure, 147, 152 reinforcer for animals, 147 relation to addiction, xxiv-xxv, 144-157 XICOT~E SELF-ADMINISTRATION PATTERIS. 161 NMOR /See also ABBREVIATIONS) metabolism in rats, 74-75 N-NITROSAMINES (See also CARCINOGENS) exposure in nontobacco products, 64, 64 in snuff, 60 in tobacco leaves, 59 metabolized by tissue, 70-75 nonvolatile, 60, 62 tobacco-specific, 60, 63 volatile, 60. 61 N-NflROSAMINES lN SMOKELESS TOBACCO, 59 NNK (See also ABBREVIATIONS) metabolism in rats, 71-73 NNN (See also ABBREVIATIONS) metabolism in rats, 73-74 relation to buccal mucosa, 73 relation to lingual mucosa, 73 NONCANCEROUS CONDITIONS (See also PRECANCEROUS CONDITIONS) relation to smokeless tobacco, 100105 NONVOLATILE NITROSAWES IN SMOIfELESS TOBACCO (PPB), 62 OFFICE ON SMOKING AND HEALTH (OSH) survey data, 7, 10, 12, 14 ORAL CANCER (See also ORAL LEUKOPLAKIA, ORAL SOFT TISSUES, LIP NEOPLASMS, MOUTH NEOPLASMS, CHEEK NEOPLASMS, AND TONGUE NEOPLASMS) case studies, 34-41 191 ORAL CANCER-Cont. epidemiologic studies, 33-55 in rats, 72, 79, 84-86, 88-89 ORAL LEUKOPLAKIA causation, vii defined, 113 prospective study, 118 relation to smokeless tobacco, viii, xxiii-xxiv, 107, 110-112, 121 studies in Scandanavia, 11 l-l 12 studies in the United States, 107, lot?-10!9, 110-112 ORAL SOFT TISSUES clinical reports of lesions, 114-117 cohort studies, 117-119 definitions, 113-l 14 oral lesions, 119-120 transformation, 113-123 PANCREATIC NEOPLASMS chewing tobacco use, 54-55 PARANASAL SINUS NEOPLASMS relation to snuff use, 48 PATHOLOGIC EFFECTS OF NICOTINE AND SMOKELESS TOBACCO coronary and peripheral vascular disease, 177-178 hypertension, 178 nonnicotine metabolic consequences, 179 peptic ulcer disease, 178 pregnancy risks, 178-179 PERIODONTAL TISSUE DEGENERATION (See also MUCOSAL PATHOLOGY, GINGIVAL AND PERIODONTAL HEALTH) and teenagers, 110 PERMIsslLE LlMIll!3 FOR INDIVWUAL N-NITROSAMINES AV CONSlIlX!TR PRODUCTS, 65 PHARMACODYNAMICS OF NICOTINE Liking Scale, 158 Morphine Benzadrine Group Scale, 158 research needs, 184 self-administration implications, 162 PHARMACOKINETICS OF NICOTINE absorption, 141-142 cotinine blood levels, 143 PHARMACOKINETICS OF NICOTINE-Cant . distribution, 142 elimination, 142-143 levels in smokeless tobacco, 141 nicotine blood levels, 143 time course of turnover, 143 PHARYNGEAL NEOPLASMS case-control study, 37,38, 39-40 cohort study, 41-42 PHYSICAL DEPENDENCE POTENTIAL OF NICOTINE (See also NEUROADAPTATION) defined, 145,162 determination, 153-154 evidence for physical dependence, 157-166 evidence of withdrawal symptoms, 164-166 implications of physical dependence, 166 reinforcement with nicotine gum, 167 studies conducted, 157, 162-163 tobacco withdrawal, 163-164 PHYSIOLOGIC EFFECTS OF NICOTINE /See also NEURO- ADAPTATION, PHYSICAL DEPENDENCE POTENTIAL OF NICOTINE) cardiovascular system, 175-l 76 central nervous system, 147,176, 183 differentiation possible, 162 endocrine system, 176 pleasant effects produced, 162, 176 PLAQUE defined, 128 use of smokeless tobacco, 130 Plug, See CHEWING TOBACCO Polonium2Ia (21OPo), See CARCINOGENS Polynuclear Aromatic Hydrocarbons (PAHI, See CARCINOGENS POSTERIOR NASAL SPACE TUMORS relation to liquid snuff, 48 PRECANCEROUS CONDITIONS (See also NONCANCEROUS CONDITIONS) Advisory Committee Report, xxi defined, 113 relation to leukoplakia, 119 relation to smokeless tobacco, xxi 192 PRECANCEROUS LESION defined, 113 oral lesions, 119 PREVXLENCE OFSMOKELESS TOBACCO USE BY CENSUS REGION, 1985,16 PREVALENCE OF USE OF SMOKE LESS TylBACCO AMONG YOUTH BYGENDERANDGRADE LOCAL SUR KEYS USING STAN- DARDIZED QUESTIONS, 21-22 PREVALENCE OF USE OF SMOKE LESS TOBACCO AMONG YOUTH BY GENDER AND GRADE.. REGIONAL AND STATE LEVEL SURVEYS REPORTED SINCE 1980,1&19 PREVALENCE OFSNUFFAND CHEWING TOBACCO USE BY ADULTM4LESlN10 GEOGRAPHIC AREAS 17 PREVALENCE OF SNUFF USE AMONG COLLEGE STUDENTS 18 YEARS OF AGE OR OLDER BY GENDER AND YEAR, 12 PREVALENCE OF THE USE OF SNUFFAND CHEWmG TOBACCO AMONG MALES BY AGE, 1970 NHLsAND1985cpssURVEY$ll Quids, See ASIAN DATA RECENCY OF SMOKELESS TOBACCO USE BY SEXAND AGE GROUP, 13 RELATmRfSK OFORAL CANCER FROMBETEL QUID W7TLjlAND W!IT.HOUT To&1CCO (WITH95 PERCENT CONFIDENCE -), 43 RELATNERLSKS OF ESOPHAGEAL CANCER lN PERSONS ExposED To CHEW- &VG TOBACCO AND SXX'i'?~ .!XlWXR Y OF M)UR CASE CONTROL STUDHi! 49 Renal Neoplasms, See URINARY TRACT NEOPLASMS RESEARCH NEEDS carcinogenesis, 93-94 nicotine exposure, 184-185 oral health effects 131-132 prevalence/trends of use, 25 standardizing methods, 25 SALIVA ANALYSIS of snuff dippers, 64 SALIVARY GLANDS effects of smokeless tobacco, lOl-l&3, 126-128 SCHEME LLNKLNG NICOTLNE TO FORMATION OF THE PROMUTAGENIC DNA ADDUCT O%METHYLGUANINE, 72 SELECTED STUDY Sm FOR THE NONC'CEROUS ORAL HEALTH EFFECTS FROM THE USE OF SMOKELESS TOBACCO, lm105 SIALADENITIS defined, 123 relation to smokeless tobacco, 127 relation to snuff dippers, 127 SIMMONS MARKET RESEARCH BUREAU survey data, 10, 12, 14-15 SMOKELESS TOBACCO (See also CHEWING TOBACCO, SNUFF) carcinogenesis associated with, 33-93 chemical constituents, 58-69 cohort studies, 41-42 commonalities with other addictive substances, 146-147, 152-157 conclusions, 24-25, 92-93, 130-131, 182-183 effect on oral leukoplakia/ mucosal pathology, 107-l 12 epidemiologic studies and case reports of oral cancer, 33-44 epidemiologic studies of other cancers, 47-55 evidence for abuse and dependence, 166-167 experimental studies exposing laboratory animals, 78-79 experimental studies of abuse and dependence, 157-166 metabolism of constituents, 70-75 nicotine addiction, 144-146 nicotine exposure, xxiv, 141-185 noncancerous and precancerous oral health effects, xxiii-xxiv, 99-130 pharmacokinetics of nicotine, 141-144 physiologic and pathologic effects, 175-l 79 prevalence and trends of use. xxii, 4-24 193 SMOKELESS TOBACCO-Cant . product characteristics, 5 research needs, 25, 93-94, 131-132, 184-185 transformation of oral soft tissue, 113-120 trends in production and sales, 5, 7 trends in self-reported use, 7, 10, 12-17, 20, 24 SMOKELESS mBACC0 AND h!EAD AND NECK CANCER BY ANA- RMfICSITE, CASECONTROL DATA FROM VtNCX!h'TAh?D MARCtlETZA, - ONLU, 38 SMO- lloBACC0 AND MOUTH CANCER CASE CONTROL DATA FROM MOOREETAL+ 36 SMO- TylBACCO AND MOUTH CANCER, CASE CONTROL DATA FROM PEACOCKETAL, 37 SNUFF (See also SMOKELESS TOBACCO, BIOASSAY8 ANIMAL MODELS) case-control studies, 37, 40 characteristics. 5 dry,7 historical uses, xviii-xix levels of carcinogens, 60, 64-66 moist, 7 relation to cancer, 41, 53-55 saliva analysis, 64 SNUFF-DIPPERS CARCINOMA defined, 113 exposure to nitrosamines, 64 oral lesions, 120 reports, 35 Scandinavian study, 111 SQUAMOUS CELL CARCINOMA case studies, 34-36 clinical study, 117 defined, 114 oral lesions, 119-120 snuff and ear neoplasms, 47 STAINING (DENTAL) defined, 129 STATE AND LOCAL SURVEY DATA adolescent use, 17, 20, 24-25 adult use, 16-17 STOMACH NEOPLASMS relation to smokeless tobacco, 51-52 Substitution Therapy, See NICOTINE ADDICTION TREATMENT SUMMRY OF DATA: LIABILJTY OF NICOTmTE AS mBACC0 SMOKE OR lNTRAVENOUS lh?JECTION$159 StJUX4RYOFREFOR~IN WZICHNICOT~ WAS AVAII ABLE uNoERINTR.AvENom DRUG SELF+tDMIhZXUATION PROC%!DiXE!$14&151 S-Y OFSELJXTZD CASE REpoRT$106 Supportive Therapy, See NICOTINE ADDICTION TREATMENT Surgeon General's Advisory Committee on the Health Consequences of Using Smokeless `Ibbacco, See ADVISORY COMMITTEE TO THE SUR- GEON GENERAL, REPORT TEETH dental caries, 129-130 effect of smokeless tobacco, 128-130 other hard tissue effects, 130 THERAPEUTIC EFFECTS OF NICOTINE as an anoretic, 154 mood regulator, 154 performance of cognitive tasks, 154 self-medication, 155 work enhancement, 154 THIRD NATIONAL CANCER STUDY (TNCS) bladder cancer, 54 laryngeal neoplasms, 50-51 other cancer sites, 55 stomach neoplasms, 52 TOBACCO SNIFFING rare practice, xvii TOBACCMPEC~IC N-NITR-B liV SMOKE LhXJ TOBACCO fPPB), 63 mBA CCO-SPECXFIC N-NITRO-Bmm' US. BRANI& 1985,66 TOBACCO WITHDRAWAL (See also PHYSICAL DEPEN- DENCE OF NICOTINE) evidence of symptoms, 164-166 194 TOBACCO WITHDRAWAL- Cont. study findings, 165 symptoms, 163-164 University of Minnesota studies, 164-166 `TONGUE NEOPLASMS (See also ORAL CANCER, MOUTH N-EOPLASMS) case-control study, 36,120 TRENDS IN PRODUCTION ANDSALES categories of products, 7 temporal trends, 7 United States Department of Agriculture (USDA) reports, 5 TRENDS IN SELF-REPORTED USE: SURVEY DATA national survey data, 7, 10, 12-15 state and local survey data, 15-1'7, 20, 24 Twist, See CHEWING TOBACCO URINARY TRACTNEOPLASMS castxontrol studies, 52-54 relationship to smokeless tobacco, 52-54 USEOFSMO-llylBAccOm THE UNITEB SZ4TEs BY VAlpIATIONs~ T- USEDm DEI?!ONS PROKiDED EylR GINGMTISAND GINGWXL RE C%ssIONBYS~~ CITED, 124 VMU4TIONS~ TERbis USEDAND D,W!MIONS PROKWED EylR USED BY STuDrn CITED, 108109 VERRUCOUS CARCINOMA defined, 114 dysplastic lesions, 13 1 voLAlzLEm-m MfOKI.hZSS mBACc0 IPPBJ, 61 WORLD HEALTH ORGANIZATION (WHO) Chnmittee on Drug Dependence, 145 defines oral leukoplakia, 107 differing diagnoses, 123 195