mm N-NITROSODIMETHYLAMINE Agency for Toxic Substances and Disease Registry US. Public Health Service — TOXICOLOGICAL PROFILE FOR N-NITROSODIMETHYLAMINE Prepared by: Syracuse Research Corporation Under Subcontract to: Clement Associates, Inc. Under Contract No. 205-88-0608 Prepared for: Agency for Toxic Substances and Disease Registry (ATSDR) U.S. Public Health Service In collaboration with U.S. Environmental Protection Agency (EPA) December 1989 paw,“ ii DISCIAIHER Mention of company name or product does not constitute endorsement by the Agency for Toxic Substances and Disease Registry. iii 53/9 ) Z “2 N32 FOREWORD T6 [/9 n 9 $79 The Superfund Amendments and Reauthorization Act of 1986 (Public F%VC%C, Law 99—499) extended and amended the Comprehensive Environmental Response, Compensation, and Liability Act of 1980 (CERCLA or Superfund). This public law (also known as SARA) directed the Agency for Toxic Substances and Disease Registry (ATSDR) to prepare toxicological profiles for hazardous substances which are most commonly found at facilities on the CERCLA National Priorities List and which pose the most significant potential threat to human health, as determined by ATSDR and the Environmental Protection Agency (EPA). The lists of the most significant hazardous substances were published in the Federal Register on April 17, 1987, and on October 20, 1988. Section 110 (3) of SARA directs the Administrator of ATSDR to prepare a toxicological profile for each substance on the list. Each profile must include the following content: (A) An examination, summary and interpretation of available toxicological information and epidemiological evaluations on the hazardous substance in order to ascertain the levels of significant human exposure for the substance and the associated acute, subacute, and chronic health effects, (B) A determination of whether adequate information on the health effects of each substance is available or in the process of development to determine levels of exposure which present a significant risk to human health of acute, subacute, or chronic health effects, and (C) Where appropriate, an identification of toxicological testing needed to identify the types or levels of exposure that may present significant risk of adverse health effects in humans. This toxicological profile is prepared in accordance with guidelines developed by ATSDR and EPA. The original guidelines were published in the Federal Register on April 17, 1987. Each profile will be revised and republished as necessary, but no less often than every 3 years, as required by SARA. The ATSDR toxicological profile is intended to characterize succinctly the toxicological and health effects information for the hazardous substance being described. Each profile identifies and reviews the key literature that iv describes a hazardous substance's toxicological properties. Other literature is presented but described in less detail than the key studies. The profile is not intended to be an exhaustive document; however, more comprehensive sources of specialty information are referenced. Each toxicological profile begins with a public health statement, which describes in nontechnical language a substance's relevant toxicological properties. Following the statement is material that presents levels of significant human exposure and, where known, significant health effects. The adequacy of information to determine a substance's health effects is described in a health effects summary. Data needs that are of significance to protection of public health will be identified by ATSDR, the National Toxicology Program of the Public Health Service, and EPA. The focus of the profiles is on health and toxicological information; therefore, we have included this information in the front of the document. The principal audiences for the toxicological profiles are health professionals at the federal, state, and local levels, interested private sector organizations and groups, and members of the public. We plan to revise these documents as additional data become available. This profile reflects our assessment of all relevant toxicological 'testing and information that has been peer reviewed. It has been reviewed by scientists from ATSDR, EPA, the Centers for Disease Control, and the National Toxicology Program. It has also been reviewed by a panel of nongovernment peer reviewers and was made available for public review. Final responsibility for the contents and views expressed in this toxicological profile resides with ATSDR. Walter R. Dowdl , Ph.D. Acting Administrator Agency for Toxic Substances and Disease Registry FOREWORD . CONTENTS LIST OF FIGURES. LIST OF TABLES . 1. PUBLIC HEALTH STATEMENT. l. l. P‘P‘H‘H‘H uvp-u:n:ra O" 7 8 WHAT IS N- NITROSODIMETHYLAMINE?. HOW MIGHT I BE EXPOSED TO N- NITROSODIMETHYLAMINE?. HOW CAN N- NITROSODIMETHYLAMINE ENTER AND LEAVE MY BODY?. HOW CAN N- NITROSODIMETHYLAMINE AFFECT MY HEALTH? . IS THERE A MEDICAL TEST TO DETERMINE WHETHER I HAVE BEEN EXPOSED TO N- NITROSODIMETHYLAMINE? . . WHAT LEVELS OF EXPOSURE HAVE RESULTED IN HARMFUL HEALTH EFFECTS? . WHAT RECOMMENDATIONS HAS THE FEDERAL GOVERNMENT MADE TO PROTECT HUMAN HEALTH?. WHERE CAN I GET MORE INFORMATlON?. 2. HEALTH EFFECTS . 2.1 INTRODUCTION . . 2. 2 DISCUSSION OF HEALTH EFFECTS BY ROUTE OF EXPOSURE. 2.2.1 Inhalation Exposure . . . . . . Death. . . Systemic Effects . Immunological Effects. Neurological Effects . Developmental Effects. Reproductive Effects . Genotoxic Effects. Cancer . posure . Death. . . Systemic Effects . Immunological Effects. Neurological Effects . Developmental Effects. Reproductive Effects . Genotoxic Effects. Cancer . 1 Exposure . Death. . Systemic Effects . Immunological Effects. Neurological Effects . Developmental Effects. Reproductive Effects . H‘P‘F‘P‘P‘H‘H H m NNNNNNNN (”\IO‘IUIJ-‘LAJNI-Jx mNG‘m‘I—‘LANH 1 E fl 0 2.2.2 . (D- . . NNNNNNHNNNNNNNN In 2.2.3 NMNNNNUNNNNNNNNONNNNNNNM wwwwwwm NNNNNNNN O‘U‘J—‘WNH iii ix xi N N H‘H‘H vi 2.2.3.7 Genotoxic Effects. 2. 2. 3. 8 Cancer . . RELEVANCE TO PUBLIC HEALTH . NM boo EFFECTS. 2.5 LEVELS IN THE ENVIRONMENT ASSOCIATED WITH LEVELS IN HUMAN TISSUES AND/0R HEALTH EFFECTS. 2.6 TOXICOKINETICS . 2.6.1 Absorption. . . 2.6.1.1 Inhalation Exposure. 2.6.1.2 Oral Exposure. 2.6.1.3 Dermal Exposure. 2.6.2 istribution. . 6.2.1 Inhalation Exposure. 6. 2. 2 Oral Exposure. .6. 2. 3 Dermal Exposure. 2.6.3 et abo lism. . . .6.3.1 Inhalation Exposure. .6. 3. 2 Oral Exposure. .6. 3. 3 Dermal Exposure. c etion . . . .6 4.1 Inhalation Exposure. 6. 4. 2 Oral Exposure. 2. 6. 4. 3 Dermal Exposure. INTERACTIONS WITH OTHER CHEMICALS. . POPULATIONS THAT ARE UNUSUALLY SUSCEPTIBLE . ADEQUACY OF THE DATABASE . . 2.9.1 Existing Information on Health Effects of N- Nitrosodimethylamine. 2.9.2 Data Needs. . 2.9.3 On-going Studies. 2.6.4 x r D 2. 2. 2 M 2 2 2 E 2 2. NNN \OCON 3. CHEMICAL AND PHYSICAL INFORMATION. 3.1 CHEMICAL IDENTITY. 3. 2 PHYSICAL AND CHEMICAL PROPERTIES 4. PRODUCTION, IMPORT, USE, AND DISPOSAL. 4.1 PRODUCTION . 4. 2 IMPORT . 4. 3 USE. 4. 4 DISPOSAL . . 4.5 ADEQUACY OF THE DATA BASE. 4. 5.1 Data Needs. 5. POTENTIAL FOR HUMAN EXPOSURE . 5.1 OVERVIEW . . . 5. 2 RELEASES TO THE ENVIRONMENT. 5.2.1 Air . 5.2.2 Water . LEVELS IN HUMAN TISSUES AND FLUIDS ASSOCIATED WITH HEALTH 39 39 39 46 47 47 47 47 47 48 48 48 48 49 49 49 49 51 51 51 51 51 51 52 52 53 53 58 59 59 59 63 63 63 63 63 64 64 65 65 66 66 66 5.3 5.4 U1UIU'I \lO‘U‘I vii 5.2.3 Soil. . . . . ENVIRONMENTAL FATE . . . . . 5.3.1 Transport and Partitioning. 5.3.2 Transformation and Degradation. 5.3.2.1 Air. 5.3.2.2 Water. 5.3.2.3 Soil . Air . Water . Soil. . . . . . . Other Media . . . . . . . . . . . . . . GENERAL POPULATION AND OCCUPATIONAL EXPOSURE . POPULATIONS WITH POTENTIALLY HIGH EXPOSURE . ADEQUACY OF THE DATABASE . 5.7.1 Data Needs. . 5.7.2 On-going Studies. UIUIUIUIF bJ—‘J-‘J—‘< ¢~uan>har ANALYTICAL METHODS 6.1 6.2 6.3 BIOLOGICAL MATERIALS . ENVIRONMENTAL SAMPLES. ADEQUACY OF THE DATABASE. 6.3.1 Data Needs. . 6.3.2 On-going Studies. REGULATIONS AND ADVISORIES . REFERENCES . GLOSSARY . APPENDIX . S MONITORED OR ESTIMATED IN THE ENVIRONMENT . 67 67 67 68 68 68 68 69 69 69 70 70 73 74 74 75 76 77 77 77 81 81 82 83 87 115 119 2-1. 2-2. 2-3. 2-4. ix LIST OF FIGURES Levels of Significant Exposure to N-Nitrosodimethylamine - Inhalation . Levels of Significant Exposure to N-Nitrosodimethylamine - Oral. Metabolism of N-Nitrosodimethylamine . Existing Information on Health Effects of N-Nitrosodimethylamine . 12 25 50 54 1-1. 1-2. 1-3. 1-4. 2-1. 2-2. 2-3. 2-4. 3-1. 3-2. 5-1. 6-1. 6-2. 7-1. xi LIST OF TABLES Human Health Effects from Breathing N-Nitrosodimethylamine Animal Health Effects from Breathing N-Nitrosodimethylamine. Human Health Effects from Eating or Drinking N—Nitroso— dimethylamine. Animal Health Effects from Eating or Drinking N-Nitroso- dimethylamine. . . Levels of Significant Exposure to N-Nitrosodimethylamine — Inhalation . Levels of Significant Exposure to N-Nitrosodimethylamine - Oral. Genotoxicity of N-Nitrosodimethylamine In Vitro. Genotoxicity of N-Nitrosodimethylamine In Vivo Chemical Identity of N-Nitrosodimethylamine. Physical and Chemical Properties of N-Nitrosodimethylamine . Detection of N-Nitrosodimethylamine in Food. Analytical Methods for Determining N-Nitrosodimethylamine in Biological Samples Analytical Methods for Determining N-Nitrosodimethylamine in Environmental Samples. Regulations and Guidelines Applicable to N-Nitrosodi- methylamine. . . 11 17 43 45 60 61 71 78 79 84 N: 6 1.134911} .3; 1. PUBLIC HEALTH STATEMENT 1.1 WHAT IS N-NITROSODIMETHYLAMINE? N-Nitrosodimethylamine is commonly known as NDMA. It is a yellow liquid which has no distinct odor. It is produced in the U.S. only for use as a research chemical. NDMA was used to make rocket fuel, but this use was stopped after unusually high levels of this compound were found in air, water, and soil samples collected near a rocket fuel manufacturing plant. NDMA is, however, unintentionally formed during various manufacturing processes at many industrial sites and in air, water and soil from reactions involving other chemicals called alkylamines. Alkylamines are both natural and man-made compounds which are found widely distributed throughout the environment. NDMA does not persist in the environment. When NDMA is released into the atmosphere, it breaks down in sunlight in a matter of minutes. When released to soil surfaces, NDMA may evaporate into air, break down upon exposure to sunlight, or sink into deeper soil. NDMA should break down within a few months in deep soil. When NDMA is released into water, it may break down upon exposure to sunlight or break down by natural biological processes. The rate of breakdown in water is not known. More information can be found in Chapters 3, 4 and 5. 1.2 HOW MIGHT I BE EXPOSED T0 N-NITROSODIMETHYLAMINE? Information suggests that the general population may be exposed to NDMA from a wide variety of sources, including environmental, consumer, and occupational sources. At this time, NDMA has been found in at least 1 out of 1177 hazardous waste sites on the National Priorities List (NFL) in the United States. Under certain conditions, NDMA may be found in outdoor air, surface waters (rivers and lakes, for example), and soil. The primary sources of human exposure to NDMA are tobacco smoke, chewing tobacco, diet [cured meats (particularly bacon), beer, fish, cheese, and other food items], toiletry and cosmetic products (for example, shampoos and cleansers), interior air of cars, and various other household goods, such as detergents and pesticides. In addition, NDMA can form in the stomach during digestion of alkylamine-containing foods. Alkylamines are naturally occurring compounds which are found in some drugs and in a variety of foods. Infants may be exposed to NDMA from the use of rubber baby bottle nipples and pacifiers which may contain very small amounts of NDMA, from ingestion of contaminated infant formulas, and from breast milk of some nursing mothers. Very low levels of NDMA have been found in some samples of human breast milk. Occupational exposure may happen in a large number of places including industries such as tanneries, pesticide manufacturing plants, rubber and tire manufacturing plants, alkylamine manufacture/use industries, fish processing industries, foundries, and dye manufacturing plants. Researchers making or handling NDMA may also be exposed to this compound if it passes through the rubber gloves they wear during laboratory work. NDMA 2 1. PUBLIC HEALTH STATEMENT has been found in groundwater samples, in amounts of 10 parts NDMA per billion parts of water, at one or more hazardous waste sites on the National Priorities List (NPL). No information is available about contamination of soil, drinking water, irrigation water, sewers, storm drains, or the human food chain with NDMA near NPL sites. For more information, refer to Chapter 5. 1.3 HOW CAN N-NITROSODIMETHYLAMINE ENTER AND LEAVE MY BODY? NDMA can enter the body when a person breathes air that contains NDMA or when a person eats food or drinks water contaminated with NDMA. NDMA can also enter the body through the skin after contact with rubber articles that contain NDMA. Experiments in animals have shown that after being given by mouth, NDMA enters the bloodstream and goes to many organs of the body in a matter of minutes. In the liver, NDMA is broken down into other substances, most of which leave the body within 24 hours in air exhaled from the lungs and in urine, along with the NDMA that is not broken down. Little is known about what happens to NDMA that enters the body through the skin or through contaminated air. Although vapors of NDMA are broken down within minutes after exposure to sunlight, if NDMA is spilled at a waste site and evaporates, a person nearby can be exposed to NDMA before it disappears from the air. The most important and probably the most harmful way of coming into contact with NDMA seems to be by eating contaminated food or drinking contaminated water. Further information on how NDMA can enter and leave the body can be found in Chapter 2. 1.4 HOW CAN N—NITROSODIMETHYLAMINE AFFECT MY HEALTH? NDMA is very harmful to the liver of animals and humans. People who were intentionally poisoned on one or several occasions with unknown levels of NDMA in beverage or food died of severe liver damage accompanied by internal bleeding. Animals that ate food, drank water, or breathed air containing high levels of NDMA over a period of days or several weeks also developed serious, non-cancerous, liver disease. When rats, mice, hamsters and other animals ate food, drank water, or breathed air containing lower levels of NDMA for periods more than several weeks, liver cancer and lung cancer as well non-cancerous liver damage occurred. The high level short- term and low level long-term exposures that caused non-cancerous liver damage and/or cancer in animals also usually resulted in internal bleeding and death. Although there are no reports of NDMA causing cancer in humans, it is reasonable to expect that exposure to NDMA by eating, drinking, or breathing could cause cancer in humans. Mice that were fed NDMA during pregnancy had offspring that were born dead or died shortly after birth. However, it is not known whether NDMA could cause the death of human babies whose mothers are exposed during pregnancy. It should be realized that exposure to NDMA does not mean that any effect on health will definitely occur. , 3 1. PUBLIC HEALTH STATEMENT 1.5 IS THERE A MEDICAL TEST TO DETERMINE WHETHER I HAVE BEEN EXPOSED TO N-NITROSODIMETHYLAMINE? The presence of NDMA can be detected in blood and urine by a test, but this test is not usually available and has not been used as a test for human exposure or to predict possible health effects. 1.6 WHAT LEVELS OF EXPOSURE HAVE RESULTED IN HARMFUL HEALTH EFFECTS? The amounts of N-nitrosodimethylamine in air, drinking water, and food that cause known health effects other than cancer in humans and animals are summarized in Tables l-l, 1-2, l-3, and 1-4. These amounts are expressed as parts of NDMA per million parts of air, water, or food (ppm). As seen in Tables 1-1 and l-3, the amounts of NDMA in air, water, or food that result in health effects in humans are unknown. As seen in Table l—2, short-term exposure of animals to air containing NDMA produces liver damage and death. Toxic effects of long-term exposures of animals to air containing NDMA are unknown. As seen in Table 1-4, short-term or long-term exposure of animals to water or food containing NDMA is also associated with serious effects, such as liver disease and death. More information on levels of NDMA that cause harmful effects in animals is presented in Chapter 2. 1.7 WHAT RECOMMENDATIONS HAS THE FEDERAL GOVERNMENT MADE TO PROTECT HUMAN HEALTH? The Federal government has issued guidelines and rules to protect human health from exposure to NDMA in water and in food. The U.S. Environmental Protection Agency (EPA) has set limits on the amounts of NDMA in water such as lakes and streams. The EPA controls the release of NDMA. Releases or spills of one pound or more of NDMA must be reported to the National Response Center. The Food and Drug Administration (FDA) has set a limit of 10 parts of NDMA per billion parts of barley malt (ppb). Further information on Federal and state regulations can be found in Chapter 7. 1.8 WHERE CAN I GET MORE INFORMATION? If you have more questions or concerns, please contact your State Health or Environmental Department or: Agency for Toxic Substances and Disease Registry Division of Toxicology 1600 Clifton Road, E-29 Atlanta, Georgia 30333 4 1. PUBLIC HEALTH STATEMENT TABLE l-l. Human Health Effects from Breathing N-Nitrosodimethylamine* Short-term Exposure (less than or equal to 14 days) Levels in Air {ppm} Length of Exposure Description of Effects The health effects resulting from short-term human exposure to air containing specific levels of NDMA are not known. Long-term Exposure (greater than 14 days) Levels in Air (ppm) Length of Exposure Description of Effects The health effects resulting from long-term human exposure to air containing specific levels of NDMA are not known. *See Section 1.2 for a discussion of exposures encountered in daily life. 5 1. PUBLIC HEALTH STATEMENT TABLE l-2. Animal Health Effects from Breathing N-Nitrosodimethylamine Short-term Exposure (less than or equal to 14 days) Levels in Air (ppm: Length pf Exposure Description of Effects* 16 4 hour Liver damage and death in dogs. Long-term Exposure (greater than 14 days) Levels in Air {ppm} Length of Exposure Description of Effects The health effects resulting from long-term animal exposure to air containing specific levels of NDMA are not known. *These effects are listed at the lowest level at which they were first observed. They may also be seen at higher levels. 6 1. PUBLIC HEALTH STATEMENT TABLE 1-3. Human Health Effects from Eating or Drinking N-Nitrosodimethylamine* Short-term Exposure (less than or equal to 14 days) Levels in Food {ppm} Length of Exposure Description of Effects The health effects resulting from short-term human exposure to food containing specific levels of NDMA are not known. Levels in Water (ppm) The health effects resulting from short-term human exposure to water containing specific levels of NDMA are not known. Long—term Exposure (greater than 14 days) Levels in Food (ppm) Len th 0 Ex 05 e sc 0 E c s The health effects resulting from long-term human exposure to food containing specific levels of NDMA are not known. Levels in Water {ppm} The health effects resulting from long-term human exposure to water containing specific levels of NDMA are not known. *See Section 1.2 for a discussion of exposures encountered in daily life. 7 1. PUBLIC HEALTH STATEMENT TABLE 1-4. Animal Health Effects from Eating or Drinking N-Nitrosodimethylamine Short-term Exposure (less than or equal to 14 days) Levels in Food {ppm} Length of Exposure Description of Effects* 75 1 week Liver damage in rats. Levels in Water gppmz 20 1 day Liver damage in hamsters. 50 1 week Death in mice. Long-term Exposure (greater than 14 days) Levels in Food gppmz Length of Exposure Description of Effects* 50 5 months Liver damage in mice. 100 62-93 days Death in rats. Levels in Water (ppm) 5.5 30 weeks Death in rats. 20 28 days Liver damage in hamsters. *These effects are listed at the lowest level at which they were first observed. They may also be seen at higher levels. 2. HEALTH EFFECTS 2.1 INTRODUCTION This chapter contains descriptions and evaluations of studies and interpretation of data on the health effects associated with exposure to NDMA. Its purpose is to present levels of significant exposure for NDMA based on toxicological studies, epidemiological investigations, and environmental exposure data. This information is presented to provide public health officials, physicians, toxicologists, and other interested individuals and groups with (1) an overall perspective of the toxicology of NDMA and (2) a depiction of significant exposure levels associated with various adverse health effects. 2.2 DISCUSSION OF HEALTH EFFECTS BY ROUTE OF EXPOSURE To help public health professionals address the needs of persons living or working near hazardous waste sites, the data in this section are organized first by route of exposure -- inhalation, oral, and dermal -- and then by health effect -- death, systemic, immunological, neurological, developmental, reproductive, genotoxic, and carcinogenic effects. These data are discussed in terms of three exposure periods -— acute, intermediate, and chronic. Levels of significant exposure for each exposure route and duration (for which data exist) are presented in tables and illustrated in figures. The points in the figures showing no-observed-adverse-effect levels (NOAELs) or lowest-observed-adverse-effect levels (LOAELs) reflect the actual doses (levels of exposure) used in the studies. LOAELs have been classified into "less serious" or "serious" effects. These distinctions are intended to help the users of the document identify the levels of exposure at which adverse health effects start to appear, determine whether or not the intensity of the effects varies with dose and/or duration, and place into perspective the possible significance of these effects to human health. The significance of the exposure levels shown on the tables and graphs may differ depending on the user's perspective. For example, physicians concerned with the interpretation of clinical findings in exposed persons or with the identification of persons with the potential to develop such disease may be interested in levels of exposure associated with "serious" effects. Public health officials and project managers concerned with response actions at Superfund sites may want information on levels of exposure associated with more subtle effects in humans or animals (LOAEL) or exposure levels below which no adverse effects (NOAEL) have been observed. Estimates of levels posing minimal risk to humans (minimal risk levels, MRLs) are of interest to health professionals and citizens alike. For certain chemicals, levels of exposure associated with carcinogenic effects may be indicated in the figures. These levels reflect the actual 10 2. HEALTH EFFECTS doses associated with the tumor incidences reported in the studies cited. Because cancer effects could occur at lower exposure levels, the figures also show estimated excess risks, ranging from a risk of one in 10,000 to one in 10,000,000 (10'4 to 10‘7), as developed by EPA. Estimates of exposure levels posing minimal risk to humans (MRLs) have been made, where data were believed reliable, for the most sensitive noncancer end point for each exposure duration. MRLs include adjustments to reflect human variability and, where appropriate, the uncertainty of extrapolating from laboratory animal data to humans. Although methods have been established to derive these levels (Barnes et a1. 1987; EPA 1980a), uncertainties are associated with the techniques. 2.2.1 Inhalation Exposure 2.2.1.1 Death At least two human deaths following inhalation of NDMA have been reported in the literature. One was a male chemist who was involved in the production of NDMA and was exposed to an unknown level of fumes for about two weeks, and subsequently to an unknown level of fumes during cleanup of a spilled flask (Freund 1937). The subject became ill 6 days later, showed abdominal distention, large amounts of yellow ascitic fluid, a tender and enlarged liver and enlarged spleen, and died 6 weeks after the last exposure. The other death was that of a male worker who was exposed to unknown concentrations of NDMA in an automobile factory. Autopsy of this subject showed a cirrhotic liver with areas of regeneration (Hamilton and Hardy 1974). The lethality of inhaled NDMA has been evaluated in several acute duration studies with animals. Four-hour single exposure LC50 values of 78 ppm (95% confidence limits of 68 and 90 ppm) and 57 ppm (95% confidence limits of 51 and 64 ppm) were determined for rats and mice, respectively (Jacobson et a1. 1955). The observation time in these assays was 14 days. The cause of death was not specified but liver damage and hemorrhage in various abdominal tissues were predominant pathologic findings. Druckrey (1967) reported that the "LDSO" for rats exposed to NDMA by inhalation for one hour is 37 mg/kg. The air concentration corresponding to this dose is not reported but a value of 925 ppm can be calculated from information provided in the report; confidence in this value is low, however, because this information is ambiguously reported. Two of three dogs that were exposed to 16 ppm NDMA for 4 hours died or were moribund by the second day (Jacobson et a1. 1955). All dogs that were similarly exposed to 43-144 ppm died or were moribund after 1-3 days. The 57 ppm mouse and 78 ppm rat LC50 values are presented in the acute duration category in Table 2-1 and Figure 2-1. The Druckrey (1967) rat value is not included in Table 2-1 and Figure 2-1 due to uncertainty regarding its validity. The 16 ppm concentration represents a LOAEL for lethality in dogs due to acute duration inhalation TABLE 2-1. Levels of Significant Exposure to N-Nitrosodimethylamine - Inhalation Exposure Graph Frequency] b LOAELa (Effect) Key Species Duration Effect NOAEL Less Serious Serious Reference (Hill) (PPR) (PM!) ACUTE EXPOSURE Death 1 rat 4 hr, 78 (LCSO) Jacobson et al. 1955 once 2 mouse 1. hr, 57 (LCSO) Jacobson et al. 1955 once 3 dog 4 hr, 16 (death) Jacobson et al. 1955 once Systemic to rat lo hr, Hepatic 78 (hemorrhagic necrosis) Jacobson et al. 1955 once 5 mouse lo hr, Hepatic 57 (hemorrhagic necrosis) Jacobson et al. 1955 once 6 dog 6 hr, Hemato 16 (increased clotting time) Jacobson et al. 1055 once 7 dog 4 hr, Hepatic 16 (hemorrhagic necrosis) Jacobson et al. 1955 once CHRONIC EXPOSURE Cancer 8 rat life, 50 (CELC) (nasal tunors) Druckrey et al. 1967 2 d/uk, 30 min/d 9 rat 25 mo, 0.07 (CEL) (liver, lmg, Moiseev and Benemanski continuous kidney tuners) 1975 10 mouse 17 mo, 0.07 (CEL) (liver, lung, Moiseev and Benemanski continuous kidney tumors) 1975 aNOAEL - No observed Adverse Effect Level OAEL - Lowest Observed Adverse Effect Level cCEL - Cancer Effect Level '3 $1033.13 Hl'IVEH II 12 2. HEALTH EFFECTS ACUTE CHRONIC (5 14 Days) (2 365 Days) .0) © '9 9 0 (08571) f 8‘“ w -_'i Ci 7a ‘. 1o . 4' 5m . .I 1 0' 0.1 ‘ . 0.01 0.001 0.0001 0.00001 0.000001 10-4 0.0000001 10“ Estimated 10 ‘ Upper-Bound 0.00000001 Human Cancer 10 _., Risk Levels 0.000000001 Key I L050 . LOAEL 10! um 011m (mum-m 0 LOAEL 101 has serious 011009 (urinals) O NOAEL (unimals) The number 00111000011 M Q CEL - emu em Lml mgmgés "‘ FIGURE 2-1. Inhalation Levels of Significant Exposure to N-Nitrosodimethylamine - 13 2. HEALTH EFFECTS exposure (Table 2-1 and Figure 2-1). The concentration of 16 ppm in air (Jacobson et a1. 1955) is presented in Table 1-2. 2.2.1.2 Systemic Effects No studies were located regarding musculoskeletal or renal effects in humans or animals following inhalation exposure to NDMA. Respiratory Effects. Freund (1937) observed small hemorrhages in the bronchi and trachea of a person who died from accidental exposure to vapors of NDMA (Section 2.2.1.1). No studies were located regarding respiratory effects in animals following inhalation exposure to NDMA. Cardiovascular Effects. Subpericardial hemorrhage was observed in a person who died from accidental exposure to vapors of NDMA (Freund 1937) (Section 2.2.1.1). No studies were located regarding cardiovascular effects in animals following inhalation exposure to NDMA. Gastrointestinal Effects. Gastrointestinal hemorrhage was observed in a person who died from accidental exposure to vapors of NDMA (Freund 1937) (Section 2.2.1.1). No studies were located regarding gastrointestinal effects in animals following inhalation exposure to NDMA. Hematological Effects. No studies were located regarding hematological effects in humans following inhalation exposure to NDMA. Hematological determinations were performed in dogs that were exposed to 16-144 ppm NDMA for 4 hours (Jacobson et a1. 1955). Increased coagulation time, increased prothrombin time, increased plasma cholinesterase levels and leukopenia occurred following exposure to all concentrations. There was no evidence of intravascular hemolysis. As indicated in Section 2.2.1.1, the concentrations producing these effects were lethal. Pathologic examination of the dogs showed bloody ascites and hemorrhage in the liver and other abdominal tissues. Due to the clinical evidence of impaired blood coagulation and the possibility that the hemorrhagic effects were related to impaired coagulation, 16 ppm is a LOAEL for hematological effects due to acute inhalation exposure (Table 2—1 and Figure 2-1). Hepatic Effects. Four cases of liver disease in humans resulting from inhalation exposure to NDMA have been described in the literature. Two of the subjects died; these cases are discussed in Section 2.2.1.1. 0f the subjects who did not die, one was a chemist who was exposed to unknown concentrations of fumes and experienced exhaustion, headache, cramps in the 14 2. HEALTH EFFECTS abdomen, soreness on the left side, nausea and vomiting for at least two years (Freund, 1937). The second case was an automobile factory worker who was exposed to unknown levels of NDMA and became violently ill with jaundice and ascites (Hamilton and Hardy 1974). Hepatotoxicity is a predominant effect of high concentrations of inhaled NDMA in animals. Pathologic examination of dogs following exposure to 16-144 ppm NDMA for 4 hours showed marked necrosis and varying degrees of hemorrhage in the liver (Jacobson et a1. 1955). Related effects at all concentrations included increased bilirubin levels and increased sulfobromophthalein retention. As indicated in Section 2.2.1.1, the concentrations producing these effects were lethal. Jacobson et al. (1955) also indicated that necrosis and hemorrhage occurred in the liver of rats and mice that were exposed to lethal concentrations of NDMA for 4 hours; as indicated in Section 2.2.1.1, LCSO values for the rats and mice are 78 and 57 ppm, respectively. The 16 ppm, 57 and 78 ppm concentrations represent LOAELs for hepatic effects due to acute inhalation exposure and are presented in Table 2-1 and Figure 2-1. The concentration of 16 ppm in air (Jacobson et a1. 1955) is presented in Table 1-2. Dermal/Ocular Effects. No studies were located regarding dermal or ocular effects in humans following inhalation exposure to NDMA. Limited information is available regarding dermal or ocular effects of inhaled NDMA. Doolittle et a1. (1984) reported that the only toxic signs observed in rats exposed to 500 or 1000 ppm for 4 hours were reddened eyes and piloerection. The only additional information reported in this study pertained to genotoxic effects. Although high concentrations of NDMA vapor are likely to be irritating, the significance of the reddened eyes and piloerection cannot be determined because it is not specified if the effects occurred at both concentrations and prevalence is not indicated. As indicated in Section 2.2.1.1, acute exposure to much lower concentrations of NDMA was lethal for rats, mice and dogs. The lack of mortality in rats at the higher concentrations in the Doolittle et a1. (1984) study may be attributable to the fact that the animals were killed immediately following exposure and consequently not observed for subsequent death. No studies were located regarding the following effects in humans or animals following inhalation exposure to NDMA: 2.2.1.3 Immunological Effects 2.2.1.4 Neurological Effects 2.2.1.5 Developmental Effects 2.2.1.6 Reproductive Effects 15 2. HEALTH EFFECTS 2.2.1.7 Genotoxic Effects No studies were located regarding genotoxic effects in humans following inhalation exposure to NDMA. Rats exposed to 500 or 1000 ppm of NDMA in the air for 4 hours showed chemically induced DNA repair in epithelial cells in the nasal turbinates and trachea. DNA repair was also evident in hepatocytes, indicating that the substance entered the general circulation. No DNA repair was seen in the pachytene spermatocytes, indicating that NDMA either did not reach the testes in high enough concentrations, or that the testes could not metabolically activate the compound (Doolittle et a1. 1984). It should be noted that the exposures in this study are likely to have been lethal if the rats had been observed following treatment; as indicated in Section 2.2.1.1, 4-hour exposure to much lower concentrations of NDMA was lethal for rats, mice and dogs in other studies. 2.2.1.8 Cancer No studies were located regarding carcinogenic effects in humans following inhalation exposure to NDMA. The carcinogenicity of inhaled NDMA has been evaluated in two studies. Twice weekly 30-minute exposures to 50 or 100 ppm NDMA vapor for life produced malignant nasal cavity tumors in rats (Druckrey et a1. 1967). The incidence of tumors was 67% in each group, and the time to induce tumors in 50% of the rats was 400 days. Group sizes were small (12 and 6 animals at 50 and 100 ppm, respectively), control data were not reported, and additional information regarding longevity was not provided. The 50 ppm concentration is included in Table 2-1 and Figure 2-1 as an effect level for cancer (cancer effect level, CEL) in rats due to intermittent inhalation exposure of chronic duration. Rats and mice that were continuously exposed to 0.07 ppm NDMA for 25 and 17 months, respectively, developed significantly increased incidences of lung, liver and kidney tumors (Moiseev and Benemanski 1975). Tumor types included various adenomas, carcinomas, and sarcomas in the lung, liver and kidneys, and hemangiomas in the liver, but the types were not tabulated according to species or concentration. Induction of nasal tumors was not reported. Exposure to 0.002 ppm NDMA according to the same schedule did not produce significantly increased incidences of tumors in either species. Since the tumors associated with exposure to 0.07 ppm NDMA are consistent with those produced by NDMA in oral and injection studies and the study is reported adequately otherwise, 0.07 ppm is considered to be a CEL for rats and mice due to continuous inhalation exposure of chronic duration (Table 2-1, Figure 2-1). EPA has adopted the oral carcinogenicity slope factor (BH) of 51 (mg/kg/day)'1 (see Section 2.2.2.8) as the slope factor for inhalation (EPA 16 2. HEALTH EFFECTS 1988a). The oral slope factor was converted to a unit risk for inhalation of 1.4 x 10'2 (pg/m3)‘l, which is equivalent to 42.4 (ppm)'l. Using this unit risk, the concentrations associated with upper bound lifetime cancer risk levels of 10-4 to 10-7 are calculated to be 2.36 x 10'6 to 2.36 x 10'9 ppm, respectively. The cancer risk levels are plotted in Figure 2-1. 2.2.2 Oral Exposure 2.2.2.1 Death At least three human deaths following oral exposure to NDMA have been reported in the literature. One of the fatalities was a woman who was apparently poisoned over a two-year period by her husband (Fussgaenger and Ditschuneit 1980, Pedal et a1. 1982). It was estimated by the authors that she received at least 4 doses as high as 250-300 mg each, for a total dose of less than 1.5 gram; the mean daily dose was estimated to be 50 pg/kg. Both clinical and autopsy findings indicated that she died of hepatic failure. Two other people (an adult male and a l-year-old boy) died within days after consuming lemonade tainted with unknown quantities of NDMA (Kimbrough 1982, Cooper and Kimbrough 1980). Based on animal studies, the authors estimated that the adult might have received about 1.3 gm, and the boy might have received about 300 mg. In both cases, clinical and autopsy findings primarily showed liver failure and cerebral hemorrhage. Single dose lethality studies have been conducted in which NDMA was administered to rats and cats by gavage. Druckrey et a1. (1967) determined a LDSO of 40 mg/kg for rats. This value was determined using an unspecified graphic technique, and confidence limits and specific mortality data were not reported. All of 12 rats that were treated with 40 mg/kg in a skin grafting (immunology) experiment died by day 21, but the stress of skin graft rejection may have contributed to mortality (Waynforth and Magee 1974). Jenkins et a1. (1985) reported that single 25 mg doses of NDMA resulted in 100% mortality in an unspecified number of rats, but it is unclear if this is dose per kg body weight or dose per total body weight. Single doses of 15 and 20 mg/kg were not lethal for nonpregnant rats but 23 mg/kg was estimated to be the LD50 for pregnant rats (Nishie 1983). The LD50 for the pregnant rats was extrapolated using mortality of 18-day pregnant rats given single oral doses of 15 or 20 mg NDMA/kg. A dose of 10 mg/kg did not produce deaths in rats within 48 hours (Sumi and Miyakawa 1983). Two of 6 cats died when treated with 50 mg NDMA/kg (Maduagwu and Basir 1980). The NOAEL and appropriate LOAEL values for lethality from these single dose studies are included in the acute duration category in Table 2-2 and Figure 2-2. The 40 mg/kg and 23 mg/kg rat LD5os are also presented in Table 2-2 and Figure 2-2. Rats, guinea pigs, cats and monkeys that were treated with NDMA by gavage at a dose of 5 mg/kg/day for 11 days experienced 30-40% mortality; TABLE 2-2. Levels of Significant Exposure to N-Nitrosodimethylamine - Oral Exposure Graph Frequency/ b LOAELC (Effect) Key Species Routea Duration Effect NOAEL Less Serious Serious Reference (mg/kslday) ACUTE EXPOSURE Death 1 rat (G) once 40 (L050) Druckrey et al. 1967 2, 3 rat (G) once 20 23 (L050) Nishie 1983 (non-pregnant rats) (pregnant rats) I. rat (G) 5-11 days, 5 (decreased Haduaguu and daily survival) Bassir 1980 S rat (G) once 10 Suni and Hiyakaua 1983 6 rat (G) 6 days, 8 (decreased McGiven and daily survival) [reton 1972 7 mouse (11) 1 uk 9.5 (decreased Terracini et al. daily survival) 1966 8 9n pig (G) 5-11 days, 5 (decreased Haduaguu and daily survival) Bassir 1980 9 hanster (H) 1, 2, lo, 7 4.0 Ungar 1984 or 14 d, daily 10 cat (G) 5-11 days, 5 (decreased Maduaguu and daily survival) Bassir 1980 11 cat (G) once 50 (death) Maduaguu and Bassi r 1980 12 monkey (G) 5-11 days, 5 (decreased Maduaguu and daily survival) Bassir 1980 'Z SLOSJJS HITVHH LI TABLE 2-2 (continued) Exposure Graph Frequency/ b LOAELC (Effect) Key Species Routea Duration Effect NOAEL Less Serious Serious Reference (mg/kg/day) Systemic 13 rat (G) once Hepatic 2.5 (degeneration) Jenkins et al. . 1985 14 rat (G) once Hepatic 20 (necrosis) Nishie 1983 15 rat (G) once Other 20 Nishie 1983 (thyroid) 16 rat (G) once Hepatic 8 (necrosis) Sumi and Miuakaua 1983 17 rat (F) 1 or 2 Hepatic 3.75 (necrosis) Khanna and Puri wk, daily 1966 18, 19 rat (G) once Hepatic 0.7 1.9 (vacuolation) Korsrud et al. 1973 20 rat (G) 5-11 d, Hepatic 5 (necrosis) Maduagwu and daily Bassir 1980 21 gn pig (G) 5-11 d, Hepatic 5 (necrosis) Maduaguu and daily Bassir 1980 22 hamster (H) 1, 2, 4, 7 Hepatic 4.0 (portal venopathy) Ungar 1984 or 14 d, daily 23 cat (G) 5-11 d, Hepatic 5 (necrosis) Maduagwu and daily Bassir 1980 24 monkey (G) 5-11 d, Hepatic 5 (necrosis) Maduaguu and daily Bassir 1980 Innmnological 25 rat (G) once 40 Haynforth and Magee 1974 'Z SlOHfifiH HL'IVHH SI TABLE 2-2 (continued) Exposure Graph Frequency/ b LOAELC (Effect) Key Species Routea Duration Effect NOAEL Less Serious Serious Reference (mg/kg/day) Developmental 26 rat (G) once, 20 (decreased Iishie 1985 G d 15, fetal weight) or 20 Cancer 27 mouse (in 1 uk, 9.5 (end) (kidney, Terracini et al. N daily luig) 1966 - INTERMEDIATE EXPOSURE :1: to > Death E 28 rat (G) 30 d, 1 Madam and daily Bassir 1980 g ’11 29, 30 rat (F) 24-110 d 2.5 5.0 (death) Barnes and Hagee g daily 1954 r—] m 31 rat (F) 40 Hk, 3.9 (decreased Hagee and Barnes v daily survival) 1956 32 rat (H) 30 wk, 0.32 (decreased Lijinsky aid 5 d/uk survival) Rater 1984 33 rat (6) 30 wk, 6.0 (decreased Lijinsky et al. 2 d/uk survival) 1987 3!. mouse (H) 49 d, 1.8 (decreased clapp and Toya daily survival) 1970 35 mouse (F) 5 mo, 5.26 (decreased Takayaa aid daily survival) Oota 1965 36 mouse (u) 13 wk, 1.9 (decreased Den Emelse daily survival) et al. 1974 6T TABLE 2-2 (continued) Exposure Graph Frequency/ b LOAELC (Effect) Key Species Routea Duration Effect NOAEL Less Serious Serious Reference (mg/kglday) 37 mouse (H) 226 d, 0.4 Clapp and Toya daily 1970 38 mouse (H) 38 wk, 1.19 (decreased Terracini et al. daily survival) 1966 39 9n pig (G) 30 d, 1 Maduaguu and daily Bassir 1980 40 hamster (G) 1. "R, 10.7 (decreased Lijinsky et al. 5° 1 d/uk survival) 1987 :l: 1.1 hamster (G) 20 wk, SJ. (decreased Lijinsky et al. I; 1 d/uk survival 1987 S 1.2 hamster (u) a, 12 or 4.0 (death) Ungar 1986 a: 16 wk, a daily '11 g 1.3 hauster (H) 28 d, 4.0 Ungar 1984 1-1 daily In 1.1. cat (6) 30 d, 1 (decreased Madam aid daily survival) Bassir 1980 45 monkey (G) 30 d, 1 NW md daily Bassir 1980 46 mink (F) 32-34 d, 0.32 (decreased Carter et al. daily survival) 1969 Systemic 1.7 rat (F) 40 wk, Hepatic 3.9 (necrosis) Hagee and Barnes daily 1956 1.8, 1.9 rat (F) 62-110 d 2.5 5.0 (necrosis) Barnes and Hagee daily 1954 OZ TABLE 2-2 (continued) Exposure Graph Frequency/ b L0AF.Lc (Effect) Key Species Routea Duration Effect NOAEL Less Serious Serious Reference (mg/kglday) 50 rat (F) 4,8 or 12 Hepatic 3.75 (necrosis) Khanna and Puri wk, daily 1966 51 rat (G) 30 d, Hepatic 1 (vacuolation) Maduaguu and daily Bassir 1980 52 mouse (F) 16-92d, Hepatic 13 (hemorrhage/ Otsuka and daily necrosis) Kuuahara 1971 53 mouse (W) 1-4 wk, Hepatic 5.0 (hemorrhage) Anderson et al. N daily 1986 '3: 54 mouse (F) 5 mo, Hepatic 5.26 (hemorrhage/ Takayana and g daily necrosis) Oota 1965 S 55 9n pig (G) 30 d, Hepatic 1 (necrosis) Madnguu and m daily Bassir 1980 a "11 56 rabbit (F) 22 wk, Hepatic 1.6 (fibrosis) Hagee and Barnes g daily 1956 I—] m 57 hamster (U) 8, 12 or Hepatic 4.0 (portal venopathy) Ungar 1986 16 wk, daily 58 hamster (H) 28 d, Hepatic 4.0 (portal venopathy) Ungar 1984 daily 59 dog (C) 3 uk, 2 d/w Hepatic 2.5 (necrosis) Stronbeck et al. (consec) 1983 60 cat (G) 30 d, Hepatic 1 (necrosis) Haduaguu and daily Bassir 1980 61 monkey (G) 30 d, Hepatic 1 (necrosis) Maduaguu and daily Bassir 1980 62 mink (F) 122 d, Hepatic 0.13 (venopathy) Koppang and daily Rimeslatten 1976 I2 TABLE 2- 2 Exposure Graph Frequency] b LOAELc (Effect) Key Species Routea Duration Effect MOAEL Less Serious Serious Reference (walks/day) 63 mink (F) 32-34 d, Hepatic 0.32 (necrosis) Carter et al. daily 1969 Developmental 64 mouse (H) 75 d, 0.02 (perinatal Anderson et al. gestation death) 1978 Cancer N 65 rat (F) 40 wk, 3.9 (CELd) (liver) Magee and Barnes daily 1956 :1: a 66 rat (u) 30 wk, 0.3 (cad) (liver) Keefer et al. [—1 S d/uk 193 g 67 rat (H) 30 wk, 0.32(CELd) (liver) Lijinsky and $1 5 d/uk Reuber 1984 '11 m 68 rat (a) 30 wk, 6.0 (cad) (liver, Lijinsky et al. 3 2 d/uk lung, kidney) 1987 m 69 mouse (H) 49 d, 1.8 (CELd) (liver, clapp and Toya daily lung) 1970 70 me (H) 38 wk, 1.19 (CELd) (liver, Terracini et al. daily luig, kidney) 1966 71 mouse (in 224 d, 0.4 (CELd) (liver) Clapp and Toya daily 1970 72 mouse (F) 10 m, 9.04 (CELd) (liver, Takayama and daily lmg) Gate 1965 7:) mouse (F) 16-92 d, 13 (CELd) (lung) Otsuka and daily Kuwahara 1971 74 mouse (G) 50 wk, 1 (CELd) (liver) Griciute et al. 2 d/Hk 1981 ZZ TABLE 2-2 (continued) Exposure Graph Frecpency/ b LOAELC (Effect) Key Species Routea Duration Effect HOAEL Less Serious Serious Reference ("Elks/day) 75 mouse (F) 5 mo, 5.26 (CELd) (liver, Takayama and daily lmg, kidney) Oota 1965 76 hamster (G) 20 wk, 5.1. (cad) (liver) Lijinsky et al. 1 d/uk 1987 77 hamster (a) 6.5 wk, 5.4 (cad) (liver) Lijinsky et al. 2 d/Hk 1987 78 hamster (G) 4 wk, 10.7 (CELd) (liver) Lijinsky et al. .N 1 d/uk 1987 d E 79 hanster (H) 12 or 4.0 (CEL ) (liver) Ungar 1986 g 16 uk, l" daily E CHRGHC EXPOSURE E: '11 Death 5 80 rat (F) 51. wk 0.5 Terao et al. (a 1978 81 mouse (H) 406 d, 0.43 (decreased Clapp and Toys daily survival) 1970 82 mink (F) 321-670 (:1 0.1 (decreased Koppang and daily survival) Rimeslatten 1976 Systemic 83 rat (F) 54 wk Hepatic 0.5 Terao et al. 1978 84 rat (F) 96 wk, Hepatic 0.5 Arai et al. 1979 daily 85 mink (F) 321-670 d Hepatic 0.1 (venopathy, Koppang and daily focal necros i s )R inmeslat ten 1976 EZ TABLE 2-2 (continued) Exposure . Graph Frquency/ b LOAELC (Effect) Key Species Routea Duration Effect NOAEL Less Serious Serious Reference (mg/kglday) Cancer 86 rat (F) 51. wk 0.5 (CELd) (testes) Terao et al. daily 1978 87 rat (F) 96 wk, 0.05 (cad) (liver) Arai et al. 1979 daily 88 rat (F) 96 wk, 10 (cud) (liver) Ito et al. 1982 daily 89 rat (in life, 0.02 (cad) (liver) Peto et al. 1981. daily 90 mouse (in life, 0.43 (cad) (liver, Clapp and Toya daily lmg) 1970 91 mink (P) 321-607 d 0.1 (cad) (liver) Koppang and daily Rimeslatten 1976 aG - savage, F - diet, ll - drinking water, C - capsule L - No Observed Adverse Effect Level cLOIlEL - Lowest Observed Adverse Effect Level dCEL - Cancer Effect Level '3 SLDHJJEI Hl’IVElH ’73 25 2 . HEALTH EFFECTS ACUTE (s 14 Days) 06.?" "9% 05$ 4? *9 5“} f 3 (mg/kg/day) 0° *9 6" ‘6‘” a» 100 7 He 25! i i ' o 3' u: 26' g o o o 2' 15' a v 10 - O . 7m ‘6: .m 6'. Go 10:13: 20,. 3'9 23¢ ‘. IICDIDC| 1". ID. " 9' I7! . g 2“ 13: 0 1w() 1 " w o 0.1 L Key r an I um In Home . LOAEL to: urban on». (calm-It) n Home! 0 LOAEL tor bu um.» oflocu (animus) g Gum: ptg O NOAEL (unlmats) mm'mn‘ough m i 32*" . CEL-Camr emu Lav-I mgmr'm‘ FIGURE 2-2. Levels of Significant Exposure to N-Nitrosodimethylamine - Oral 26 2 . HEALTH EFFECTS INTERMEDIATE (15 - 364 Days) 5 4: '5 (mg/kg/day) of Q39 f of 100 [- 52m 73m 0 O m 79: 10 _ . . 72m 9 33' 54m 0 35m 41. 40' ".7” 7077 3° . 8 m 5!» 57.9- . z n.- ' g: m ‘ so: . m '5' O m 39' ‘g 4. 1— 0"”.0 oo o 09».- "W a: 30m «c 51: 55¢ 00: m 74m 37m can or: . 7m - O .z ' .2 32: 0.1 -, 52 um I 0.01 — Key I an . LOAEvamIous cumsmlmm) m Mouo o LOAEvahu subu- otlocllhnlmnls) h Rabbi! O NOAEL(InImaIs) I Ham-m . GEL-CancerEfloa owl Trumanmxuquch n! g Guhn P69 canspcndgtoommsln a Dog accompanying tab“. c cm I: Manny n Mink FIGURE 2 - 2 (continued) 27 2 . HEALTH EFFECTS CEL - CIDCOI Efloc! Laval Thommbormxnouch m CHRONIC (a 365 Days) 4%" 3 (mg/kg/day) of $3 0" 10 - 9.. 1 — so: am 03.04: w m 0 O O 0.1 _ . l2n 3;" '7' . m 0.01 - to: 0.001 -- 0.0001 — 0.00001 _ 0.000001 — 10.. 0.0000001 - 10-5 Estimated _ Upper-Bound o.ooooooo1 _ ‘°‘ Human Cancer oooooooom 10‘7 “is“m’e's Key 7 Rat . LOAEL bf «riots 0110c: (annuals) m Mouu o LOAEL for loss urbus 0110c: (animals) n max 0 NOAEL(InImaIs) O 10 b1 mmmam'sn W FIGURE 2-2 (continued) 28 2. HEALTH EFFECTS mean survival times were 5-11 days (Maduagwu and Bassir 1980). Rats treated by gavage daily with 8 mg/kg NDMA for 6 days experienced 10% mortality within one month (McGiven and Ireton 1972). Administration of NDMA in the drinking water at a daily dose of 9.5 mg/kg for one week resulted in decreased survival in mice (Terracini et al. 1966). Administration of a daily dose of 4 mg/kg/day in the drinking water of hamsters for l, 2, 4, 7 or 14 days did not result in mortality (Ungar 1984). The hamster NOAEL value and all LOAEL values for lethality from these repeated exposure studies are recorded in Table 2-2 and plotted in Figure 2-2. The mouse dose of 9.5 mg/kg/day was calculated from the administered concentration of 50 ppm in water (Terracini et a1. 1966); this concentration is presented in Table 1-4 for short-term exposure. Numerous oral studies in which NDMA was administered for intermediate durations (15-365 days) have been conducted. Deaths resulting from intermediate duration exposure to NDMA were usually attributed to liver toxicity or carcinogenicity. Representative lethal and nonlethal intermediate duration exposures in various species are presented below. In rats, decreased survival resulted when 0.32 mg NDMA/kg was given in the drinking water for 5 days/week for 30 weeks (Lijinsky and Reuber 1984), and when 6 mg/kg was administered by gavage for 2 days/week for 30 weeks (Lijinsky et al. 1987). Control groups were not included in the latter study but there was 100% mortality by 40 weeks after cessation of treatment. Barnes and Magee (1954) administered NDMA in the diet to small numbers of rats (6/group); 2.5 mg/kg/day produced no deaths, 5 mg/kg/day produced 100% mortality after 62-93 days, and 10 mg/kg/day produced 100% mortality after 34-37 days. Rats treated with 3.9 mg/kg/day in the diet for 40 weeks also had high mortality (Magee and Barnes 1956). Daily exposure to 1 mg/kg/day by gavage for 30 days had no effect on survival of rats (Maduagwu and Bassir 1980). Jenkins et al. (1985) observed mortality in rats that received 2.5 mg doses of NDMA by gavage for 4 days/week for 9 weeks, but it is unclear if this is dose per kg body weight or dose per total body weight. The NOAEL values and all reliable LOAEL values for lethality in rats from these intermediate duration studies are recorded in Table 2—2 and plotted in Figure 2-2. The rat dose of 5 mg/kg/day was calculated from the administered concentration of 100 ppm in diet (Barnes and Magee 1954); this concentration is presented in Table 1-4 for long-term exposure. The rat dose of 0.32 mg/kg/day was calculated from the administered concentration of 5.5 ppm in water (Lijinsky and Reuber 1984); this concentration is also presented in Table 1-4 for long-term exposure. In intermediate duration studies with mice, decreased survival resulted from treatment with doses of 1.8 mg/kg/day via drinking water for 49 days (Clapp and Toya 1970), 1.9 mg/kg/day via drinking water for 13 weeks (Den Engelse et a1. 1974), 1.19 mg/kg/day via drinking water for 38 weeks (Terracini et a1. 1966) and 5.26 mg/kg/day via diet for 5 months (Takayama and Oota 1965). Mice that received 0.4 mg/kg/day in drinking water for 224 days did not experience significantly decreased survival (Clapp and Toya 29 2. HEALTH EFFECTS 1970). The NOAEL value and all LOAEL values for lethality in mice from these intermediate duration studies are recorded in Table 2-2 and plotted in Figure 2-2. Survival data for intermediate oral exposure to NDMA are available for species other than rat and mouse. Daily gavage exposure to 1 mg NDMA/kg for 30 days caused decreased survival in cats but not guinea pigs or monkeys (Maduagwu and Bassir 1980). In hamsters, daily administration of 4 mg/kg/day in the drinking water for 8, 12, or 16 weeks resulted in occasional moribundity (Ungar 1986), while no lethality resulted from daily administration of the same dose for 28 days (Ungar 1984); this dose is a NOAEL or LOAEL for lethality depending on duration of exposure. Once weekly gavage treatment with a dose of 10.7 mg/kg for 4 weeks or 5.4 mg/kg for 20 weeks was lethal for hamsters (Lijinsky et a1. 1987). Mink that were given doses of 0.32 or 0.63 mg/kg/day in the diet died after 23-34 days of treatment (Carter et al. 1969), but low numbers of animals were tested (three per dose). Mink fed a contaminated diet that provided approximately 0.18 mg NDMA/kg/day died (Martino et a1. 1988), but there is uncertainty about the dietary concentration of NDMA used to calculate the dose. The mink that were examined in this study were from a commercial breeding colony that died during a 2 month period; durations of exposure were not specified. The NOAEL values and all reliable LOAEL values for lethality in these intermediate duration studies are recorded in Table 2-2 and plotted in Figure 2-2. Chronic lethality data are available for NDMA-exposed rats, mice and mink. Survival of rats that received 0.5 mg/kg/day of NDMA in the diet for 54 weeks (Terao et a1. 1978) was not affected. Decreased survival occurred in mice that were exposed to 0.43 mg/kg/day in the drinking water for life (average 406 days) (Clapp and Toya 1970). Mink appear to be particularly sensitive to NDMA as mortality resulted from ingestion of 0.1 mg/kg/day in the diet for 321-670 days (Koppang and Rimeslatten 1976). The NOAEL value and LOAEL values for lethality in these chronic duration studies are recorded in Table 2-2 and plotted in Figure 2-2. 2.2.2.2 Systemic Effects No studies were located regarding hematological, musculoskeletal or dermal/ocular effects in humans or animals following oral exposure to NDMA. Respiratory Effects. Petechial and larger hemorrhages were observed in the lungs of two people following lethal poisoning with NDMA (Kimbrough 1982) (Section 2.2.2.1). Macroscopic congestion was noted in the lungs of rats that were administered 3.75 mg/kg/day doses of NDMA in the diet for 1-12 weeks (Khanna and Puri 1966). The adversity of the congestion cannot be determined because results of lung histological examinations were not reported. No studies were 30 2. HEALTH EFFECTS located regarding respiratory effects in animals due to chronic duration oral exposure. Cardiovascular Effects. Myocardial and endocardial bleeding was observed in a person following lethal poisoning with NDMA (Kimbrough 1982) (Section 2.2.2.1). Macroscopic congestion was noted in the myocardium of rats that were administered 3.75 mg/kg/day doses of NDMA in the diet for l-12 weeks (Khanna and Puri 1966). The adversity of the congestion cannot be determined because results of heart histological examinations were not reported. No studies were located regarding cardiovascular effects in animals due to chronic duration exposure. Gastrointestinal Effects. Gastrointestinal hemorrhage occurred in humans following lethal poisoning with NDMA (Kimbrough 1982, Pedal et a1. 1982) (Section 2.2.2.1). NDMA produced similar gastrointestinal effects in animals. Barnes and Magee (1954) observed occasional hemorrhage into the gastrointestinal tract in rats that died from treatment with a single 50 mg/kg dose of NDMA by gavage, or with 10 mg/kg/day doses in the diet for 34-37 days. The numbers of animals examined were unspecified (single dose study) or small (6 in the diet study), and frequency of occurrence was not indicated. Gastrointestinal hemorrhages were also observed in mink that ingested 0.32 or 0.63 mg NDMA/kg/day via diet for 23-34 days (Carter et a1. 1969). Only three mink L per dose were treated, the hemorrhages occurred in a total of three mink, and the dose(s) that the affected mink received was not specified. The cause of the hemorrhages in the mink was attributed to gastric and duodenal erosions. No studies were located regarding gastrointestinal effects in animals due to chronic duration exposure. Hepatic Effects. Five members of a family who consumed unknown quantities of NDMA in lemonade became ill with nausea and vomiting associated with acute liver disease, generalized bleeding and low platelet counts (Kimbrough 1982, Cooper and Kimbrough 1980). As indicated in Section 2.2.2.1, two of these people died; the other three were released from a hospital 4-21 days after admission. Another fatality due to ingestion of NDMA was attributed to liver failure (Fussgaenger and Ditschuneit 1980, Pedal et al. 1982) (Section 2.2.2.1). Autopsies of the subjects described above showed that the primary effects were hemorrhagic and cirrhotic changes in the liver and necrosis and hemorrhage in other internal organs. Hepatotoxicity of NDMA has been described and investigated in numerous oral studies of acute, intermediate and chronic duration in several animal species. Hepatotoxicity is the most prominent and characteristic systemic effect of NDMA, resulting in centrilobular necrosis and hemorrhage often leading to hemorrhagic ascites. 31 2. HEALTH EFFECTS In acute studies, characteristic hepatotoxic alterations, as indicated above, occurred in rats following single gavage doses as low as 20 and 8 mg/kg (Nishie 1983, Sumi and Miyakawa 1983), and following daily doses of 3.75 mg/kg in the diet for 1 or 2 weeks (Khanna and Puri 1966). These doses therefore are LOAELs for serious hepatic effects. Jenkins et a1. (1985) observed degenerative alterations in the livers of rats following a single 2.5 mg/kg gavage dose of NDMA. As these alterations (collapse of reticulum network in the centrilobular areas followed by regeneration) were nonnecrotic and did not result in loss of the lobular architecture, the 2.5 mg/kg dose is a LOAEL for less serious hepatic effects. Single gavage doses of 1.9 mg/kg and 0.7 mg/kg are a LOAEL for less serious hepatic effects and a NOAEL, respectively, for rats, as nonnecrotic histologic alterations (clumping and slight vacuolation of cells in the central vein area) occurred at 1.9 mg/kg and no alterations occurred at 0.7 mg/kg (Korsrud et a1. 1973). Daily gavage exposure to 5 mg/kg for 5-11 days produced hemorrhagic necrosis in rats, guinea pigs, cats and monkeys (Maduagwu and Bassir 1980). Hamsters that ingested daily doses of 4 mg/kg/day in the drinking water for 1, 2, 4, 7 or 14 days showed portal venopathy, a less serious hepatic effect (Ungar 1984). The NOAEL value and LOAEL values for hepatic effects in these acute duration studies are recorded in Table 2-2 and plotted in Figure 2-2. The rat diet dose of 3.75 mg/kg/day was calculated from the administered concentration of 75 ppm in food (Khanna and Puri 1966); this concentration is presented in Table 1-4 for short-term exposure. The hamster drinking water dose of 4 mg/kg/day was calculated from the administered concentration of 20 ppm in water (Ungar 1984); this concentration is also presented in Table 1-4 for short-term exposure. In intermediate duration studies with rats, characteristic hepatic effects (described previously) were produced by treatment with NDMA doses of 3.75 mg/kg/day in the diet for 4-12 weeks (Khanna and Puri 1966), 5 mg/kg/day in the diet for 62—95 days (Barnes and Magee 1954), and 3.9 mg/kg/day in the diet for 40 weeks (Magee and Barnes 1956). Jenkins et al. (1985) observed cirrhosis in rats that received 2.5 mg doses of NDMA by gavage for 4 days/week for 9 weeks, but it is unclear if this is dose per kg body weight or dose per total body weight. A dose of 1 mg/kg/day administered by gavage for 30 days produced centrilobular congestion and vacuolation of hepatocytes without necrosis in rats (Maduagwu and Bassir 1980), indicating that this dose is a LOAEL for less serious hepatic effects. Hepatic alterations were not observed in rats treated with 2.5 mg/kg/day in the diet for 110 days (Barnes and Magee 1954). The NOAEL and all LOAEL values for hepatic effects in rats from these intermediate duration studies are recorded in Table 2-2 and plotted in Figure 2-2. Characteristic liver alterations (described previously) occurred in mice that were treated with NDMA doses of 5.0 mg/kg/day in the drinking water for 1-4 weeks (Anderson et a1. 1986), 13 mg/kg/day in the diet for 16- 92 days (Otsuka and Kuwahara 1971) and 5.26 mg/kg/day in the diet for 5 months (Takayama and Oota 1965). These LOAELs for hepatic effects in mice due to intermediate duration exposure are included in Table 2-2 and plotted 32 2. HEALTH EFFECTS in Figure 2-2. The mouse dose of 5.26 mg/kg/day was calculated from the administered concentration of 50 ppm in food (Takayama and Oota 1965); this concentration is presented in Table 1-4 for long-term exposure. Liver effects resulting from intermediate duration oral exposure have been observed in species other than rat and mouse. Treatment with 1 mg/kg/day by gavage for 30 days was hepatotoxic for guinea pigs, cats and monkeys (Maduagwa and Basir 1980). Necrotic alterations occurred in dogs treated with 2.5 mg/kg by capsule on 2 days/week for 3 weeks (Strombeck et a1. 1983). Fibrotic and proliferative alterations without necrosis or hemorrhage were observed in rabbits treated with an average NDMA dose of 1.6 mg/kg/day in the diet for 22 weeks (Magee and Barnes 1956), indicating that this dose is a less serious LOAEL for hepatic effects. Occlusive alterations in the portal veins developed in hamsters that received daily 4 mg/kg doses in the drinking water for 28 days or 8, 12 or 16 weeks (Ungar 1984, 1986), indicating that this dose is also a less serious LOAEL for hepatic effects. Similar hepatic venopathy occurred in mink exposed to 0.13-0.15 mg/kg/day in the diet for 122 days (Koppang and Rimeslatten 1976). Mink that were given doses of 0.32 or 0.63 mg/kg/day in the diet for 23-34 days had widespread liver necrosis (Carter et a1. 1969), but low numbers of animals were tested (three per dose). Liver necrosis was also observed in mink that ingested 0.18 mg/kg/day via diet (Martino et a1. 1988); limitations of this study, discussed in Section 2.2.2.1, include uncertainty regarding exposure duration and concentration. All reliable LOAEL values for hepatic effects due to intermediate duration exposure in these studies are recorded in Table 2-2 and plotted in Figure 2-2. The hamster drinking water dose of 4 mg/kg/day was calculated from the administered concentration of 20 ppm in water (Ungar 1984, 1986); this concentration is presented in Table 1-4 for long-term exposure. It should be noted that this water concentration (20 ppm) is higher than the water concentration associated with death (5.5 ppm) due to long-term exposure reported in Table 1-4. The apparent discrepancy in these values is attributable to differences in species sensitivity and length of exposure (rats exposed for 30 weeks, hamsters exposed for 28 days). In chronic duration studies, characteristic hepatotoxic alterations (described previously) were not observed in rats that were treated with 0.5 mg/kg/day NDMA in the diet for 54 weeks (Terao et a1. 1978) or 96 weeks (Arai et a1. 1979). Alterations in mink that ingested 0.1 mg/kg/day doses of NDMA in the diet for 321-670 days included occlusive changes in the hepatic veins with focal necrosis (Koppang and Rimeslatten 1976). Data regarding hepatic effects of chronic oral NDMA exposure in other species were not found in the available literature. The NOAEL values and LOAEL value for hepatic effects due to chronic exposure in these studies are recorded in Table 2-2 and plotted in Figure 2-2. Although hepatotoxicity is the primary effect of NDMA and has been demonstrated in all tested species, calculation of MRLs for NDMA is precluded by insufficient data defining the threshold region (i.e., NOAELs) 33 2. HEALTH EFFECTS for intermediate and chronic exposures, particularly for species which appear to be particularly sensitive (e.g., mink) and because serious effects (perinatal death) occurred in a developmental study (see Section 2.2.2.5) at a dose lower than any NOAELs for liver effects. Renal Effects. No studies were located regarding renal effects in humans following oral exposure to NDMA. Limited information is available regarding renal effects of orally- administered NDMA in animals. In a study by Nishie (1983), pregnant and nonpregnant rats were treated with a single NDMA dose of 15 or 20 mg/kg/day by gavage. An unspecified number of deceased animals (dose and pregnancy state not indicated) had distal tubule necrosis two days following treatment, and surviving rats had normal kidneys. Macroscopic congestion was noted in kidneys of rats that were administered 3.75 mg/kg/day doses of NDMA in the diet for 1-12 weeks (Khanna and Puri 1966). The adversity of the congestion cannot be determined because results of kidney histological examinations were not reported. Moderate tubule congestion with other effects (glomerulus dilatation, slightly thickened Bowman's capsule) were observed in mink that ingested 0.18 mg/kg/day via diet (Martino et a1. 1988); limitations of this study, discussed in Section 2.2.2.1, include uncertainty regarding exposure duration and concentration. Other Systemic Effects. Adrenal relative weight and mitotic count were increased in rats following a single 20 mg/kg gavage dose of NDMA (Nishie et a1. 1983). Other results of the adrenal histological examinations were not described, precluding assessment of adversity of the increased adrenal weight. There was no effect on thyroid weight or histology in the same study. It therefore is appropriate to regard 20 mg/kg as a NOAEL for thyroid effects in rats due to acute oral exposure (Table 2-2 and Figure 2-2). Macroscopic congestion was noted in spleens of rats that were administered 3.75 mg/kg/day doses of NDMA in the diet for 1-12 weeks (Khanna and Puri 1966). The adversity of the congestion cannot be determined because results of spleen histological examinations were not reported. 2.2.2.3 Immunological Effects No studies were located regarding immunological effects in humans following oral exposure to NDMA. Limited information is available regarding immunological effects of orally-administered NDMA in animals. Skin graft survival time and white blood cell count were not reduced in rats that received a single 40 mg/kg dose of NDMA by gavage, indicating that treatment was not immunosuppressive (Waynforth and Magee 1974). The dose reported was near the LD50 for rats, but all of the animals died by day 21; it is indicated that the high mortality may partially reflect the stress of skin graft rejection. Although treatment resulted in 100% mortality, this dose represents a NOAEL for immunological effects due to acute duration oral exposure (Table 2-2 and 34 2. HEALTH EFFECTS Figure 2-2). No studies were located regarding immunological effects in animals following intermediate or chronic duration exposure to NDMA. 2.2.2.4 Neurological Effects No studies were located regarding neurological effects in humans following oral exposure to NDMA. Dogs treated with 2.5 mg NDMA/kg/day by capsule on 2 consecutive days/week for 3 weeks experienced marked central nervous system (CNS) depression (Strombeck et a1. 1983). The significance of this observation cannot be ascertained since it was not characterized further. As these dogs developed liver necrosis and hepatic insufficiency, it is possible that the CNS depression is secondary to liver damage rather than a direct neurological effect of NDMA. 2.2.2.5 Developmental Effects No studies were located regarding developmental effects in humans following oral exposure to NDMA. Evidence indicates that orally-administered NDMA is a developmental toxicant in animals. Fetuses of rats that received single 20 mg/kg doses of NDMA by gavage on days 15 or 20 of gestation had significantly decreased body weights, but fetal survival data were not reported (Nishie 1983). This dose was also toxic to the dams as indicated by reduced body weight, hepatotoxicity and mortality. Other investigators have reported fetal mortality in rats that were treated with a single 30 mg/kg dose of NDMA by gavage on various days during the first 12 days (Aleksandrov 1974) or 15 days (Napalkov and Alexandrov 1968) of gestation. In other studies, NDMA reportedly caused fetal deaths in rats when administered in the diet at a dose of 5 mg/kg/day from an unspecified day in early pregnancy (treatment duration not indicated) (Bhattacharyya 1965), by gavage at a dose of 2.9 mg/kg/day during the first or second weeks of gestation (Napalkov and Alekandrov 1968), or by gavage at a dose of 1.4 mg/kg/day throughout gestation until days 17-21 (not specified) (Napalkov and Alekandrov 1968). Teratogenic effects were not observed in the studies of Aleksandrov (1974) and Napalkov and Alekandrov (1968), and not evaluated in the studies of Nishie (1983) and Bhattacharyya (1965). Evaluation of the studies of Bhattacharyya (1965), Napalkov and Alekandrov (1968) and Aleksandrov (1974) is complicated by insufficient information regarding experimental design and results; deficiencies include lack of control data, lack of maternal toxicity data, use of pooled data and/or uncertain treatment schedule. Due to these limitations, there is low confidence in the doses associated with fetotoxicity in these studies. As Nishie (1983) is the only adequately reported fetotoxicity study, 20 mg/kg is presented as a LOAEL for developmental effects in rats due to acute exposure to NDMA in Table 2-2 and Figure 2-2. 35 2. HEALTH EFFECTS In another experiment conducted by Aleksandrov (1974), a single dose of 30 mg NDMA/kg was administered by gavage to rats on day 21 of gestation. Histological examination of the offspring at the time of natural death (2274 days after exposure) reportedly showed tumors in 5 of 20 animals. Although this is possibly a manifestation of transplacental carcinogenesis, evaluation of this finding is precluded by limitations including a lack of control data and inadequate reporting of tumor types. Increased perinatal mortality (stillbirths and newborn deaths) occurred in mice as a consequence of maternal treatment with 0.02 mg NDMA/kg/day in the drinking water (Anderson et al. 1978). The mice were treated for 75 days prior to mating and throughout pregnancy and lactation. Histological examinations of the stillborn fetuses and dead neonates showed no abnormalities. The 0.02 mg/kg/day dose represents a LOAEL for developmental effects due to intermediate duration exposure (Table 2-2 and Figure 2-2). 2.2.2.6 Reproductive Effects No studies were located regarding reproductive effects in humans following oral exposure to NDMA. There was no significant increase in time-to-conception in mice that were exposed to 0.02 mg NDMA/kg/day via drinking water for 75 days prior to mating (Anderson et a1. 1978). Other reproductive indices were not evaluated. 2.2.2.7 Genotoxic Effects Methylated DNA (7-methy1guanine and 06-methy1guanine) was detected in the liver of a victim of suspected NDMA poisoning (Herron and Shank 1980). Additional studies regarding genotoxic effects in humans following oral exposure to NDMA were not located. Oral studies with rats indicate that the liver is sensitive to the genotoxic effects of NDMA. When administered by gavage at a dose of 5.2 mg/kg, NDMA induced damage in rat liver DNA as measured by increased alkaline elution (Brambilla et a1. 1981). When administered to rats via diet at a dose of 2.5 mg/kg/day, NDMA induced DNA damage in the liver as measured by a slow sedimentation in alkaline sucrose gradients (Abanobi et a1. 1979). The effect was first observed after 2 days of feeding, and became progressively worse during the next 8 weeks of feeding; no proportionate increases in damage occurred when the feedings were continued for 15 or 31 weeks. DNA synthesis and repair was detected in the liver of rats treated with single 10 or 50 mg/kg doses by gavage (Bermudez et al. 1982). Radiolabeled thymidine uptake during mouse testicular DNA synthesis was inhibited by a single gavage dose of 50 mg NDMA/kg (Friedman and Staub 1976). 36 2. HEALTH EFFECTS Administration of NDMA to hamsters by gavage at doses of 50, 100, or 200 mg/kg on the 11th or 12th day of pregnancy caused micronucleus formation and chromosomal aberrations in the embryonic fibroblasts (Inui et al. 1979). NDMA did not induce significant increases in sister chromatid exchanges in bone marrow cells of Chinese hamsters following gavage administration of 12.5-400 mg/kg (Neal and Probst 1983). 2.2.2.8 Cancer No studies were located regarding carcinogenic effects in humans following oral exposure to NDMA. The carcinogenicity of orally-administered NDMA has been demonstrated unequivocally in acute, intermediate and chronic duration studies with rats, mice, hamsters and mink. The liver and lungs are the primary targets for NDMA carcinogenesis but tumors of the kidneys and testes can also occur. Incidences of liver and lung tumors are generally very high (often 50-100Z), but liver tumors appear to occur most frequently in rats and hamsters and lung tumors appear to occur most frequently in mice. The liver tumors are usually hemangiosarcomas and hepatocellular carcinomas, and lung tumors are usually adenomas and liver tumor metastases. Low incidences of epithelial tumors (8.6%) and mesenchymal tumors (14.5%) developed in the kidneys of rats following treatment with 8 mg NDMA/kg/day for 6 days (McGiven and Ireton 1972, Ireton et a1. 1972). Evaluation of these data is complicated by the lack of a control group. Daily diet treatment with 9.5 mg/kg for one week produced kidney and lung adenomas in mice (Terracini et al. 1966). No other acute duration oral carcinogenicity studies were found in the reviewed literature. The CEL from the mouse study is presented in the acute duration category in Table 2-2 and in Figure 2-2. Numerous oral carcinogenicity studies of NDMA of intermediate duration have been conducted. Treatment durations were often in the range of 20-40 weeks, frequency of treatment ranged from once weekly to daily, and carcinogenicity was observed in all studies. Studies representing various treatment durations and various methods of oral treatment (drinking water, diet and gavage) for the lowest doses in different species are identified below. Rats administered NDMA in the drinking water at doses of 0.3 mg/kg/day, 5 days/week for 30 weeks, developed malignant liver tumors (Keefer et a1. 1973, Lijinsky and Reuber 1984). Lijinsky et al. (1987) observed high incidences of liver, lung and kidney tumors in rats that were treated by gavage with 6 mg NDMA/kg twice weekly for 30 weeks; controls were not used in this study. In an intermediate duration diet study with rats, daily treatment with a dose of 3.9 mg/kg for 40 weeks resulted in a 95% incidence of hepatic tumors (Magee and Barnes 1956). The CELs from these 37 2. HEALTH EFFECTS intermediate duration studies with rats are recorded in Table 2~2 and plotted in Figure 2-2. Liver, lung and/or kidney tumors developed in mice that were exposed to NDMA daily via drinking water at doses of 1.8 mg/kg for 49 days (Clapp and Toya 1970), 1.19 mg/kg for 38 weeks (Terracini et a1. 1966) and 0.4 mg/kg for 224 days (Clapp and Toya 1970). Daily administration of NDMA via diet at doses of 13 mg/kg for 16-92 days (Otsuka and Kuwahara 1971), 5.26 mg/kg for 5 months (Takayama and Oota 1965) and 9.04 mg/kg for 10 months (Takayama and Oota 1965) also induced liver, lung and/or kidney tumors in mice. In the only intermediate duration gavage study with mice, twice weekly doses of 1 mg/kg for 50 weeks resulted in high (37-53%) incidences of malignant liver tumors (Griciute et a1. 1981). The CELs from these intermediate duration studies with mice are recorded in Table 2-2 and plotted in Figure 2—2. Hamsters that were treated with NDMA by gavage twice weekly with a dose of 5.4 mg/kg for 6.5 weeks, once weekly with a dose of 10.7 mg/kg for 4 weeks, or once weekly with a dose of 5.4 mg/kg for 20 weeks developed high (60-79%) incidences of liver tumors (Lijinsky et a1. 1987). However, control groups were not included in the study of Lijinsky et a1. (1987). Daily administration of 4 mg/kg in the drinking water to hamsters for 12 or 16 weeks resulted in high incidences of cholangiocellular adenocarcinomas (Ungar 1986). The CELs from these intermediate duration studies with hamsters are recorded in Table 2-2 and plotted in Figure 2-2. Hemangiomatous liver tumors occurred in 55% of deceased mink that received NDMA in the diet at an estimated dose of 0.18 mg/kg/day (Martino et a1. 1988); limitations of this study, discussed in Section 2.2.2.1, include uncertainty regarding exposure duration and concentration, examination only of animals that died and use of historical controls. Due to the limitations of this study, it is inappropriate to present a CEL for mink due to intermediate duration exposure in Table 2-2 and Figure 2-2. Chronic oral carcinogenicity studies of NDMA have been conducted with rats, mice and mink. Tumors at sites other than the liver and testis have not been associated with chronic treatment. Terao et al. (1978) observed a 47% increase in the incidence of testicular Leydig-cell tumors, but no tumors in the liver or other tissues, in rats that were treated with 0.5 mg/kg daily doses of NDMA in the diet for 54 weeks. Increased incidences of liver tumors, but not testicular interstitial cell tumors, occurred in rats that received 0.05 or 0.5 mg/kg/day doses of NDMA in the diet for 96 weeks (Arai et a1. 1979). In this study, liver tumor incidences were generally higher in female rats than in male rats. Increased incidences of liver tumors also occurred in rats that were treated with NDMA in the diet for 96 weeks at a dose of 10.0 mg/kg/day (Ito et a1. 1982); similar treatment with doses of 0.1 or 1.0 mg/kg/day did not produce increased incidences of liver tumors. It should be noted that Wistar rats were tested in both the Ito et a1. (1982) and Arai et al. (1979) studies. The reason for the lack of liver tumors at doses below the relatively high 10 mg/kg/day dose in the Ito et a1. (1979) study is not clear, but may be related to low susceptibility of 38 2. HEALTH EFFECTS male rats. In a lifetime drinking water study, Peto et a1. (1984) administered doses of 0.001-1 2 mg/kg/day to rats and observed that incidences of liver tumors were significantly increased at 20.02 mg/kg/day; median survival time at the lowest tumorigenic doses was in the range of 28- 31 months. Crampton (1980) administered NDMA to rats in the drinking water at doses ranging from 0.002-l.5 mg/kg/day for life and observed increased liver tumor incidences at 20.008 mg/kg/day; median survival time at 0.008 mg/kg day was >900 days. The results reported by Crampton (1980) were preliminary and there is uncertainty regarding the dosages; ppm concentrations in water and mg/kg/day equivalency were reported, but the basis for the equivalency is not indicated and the conversion cannot be verified using standard methodology. Clapp and Toya (1970) administered NDMA to mice via drinking water at daily doses of 0.43 and 0.91 mg/kg/day for life and observed that incidences of lung tumors and liver hemangiosarcomas were significantly increased at both doses; mean survival time at the low and high doses were 12 and 17 months, respectively. Hemangiomatous liver tumors developed in mink exposed to 0.1 mg/kg/day NDMA in the diet for 321- 607 days (Koppang and Rimeslatten 1976). The CELs for rats, mice and mink from these chronic studies, except the uncertain value from the Crampton (1980) study, are recorded in Table 2—2 and plotted in Figure 2-2. The EPA (1988a) has derived and verified an oral slope factor (BH) of 51 (mg/kg/day)'1 for NDMA based on the liver tumor response in the Peto et a1. (1984) study. Using this slope factor, the doses associated with upper bound lifetime cancer risk levels of 10'4 to 10'7 are calculated to be 1.96 x 10‘6 to 1.96 x 10'9 mg/kg/day, respectively. The cancer risk levels are plotted in Figure 2-2. 2.2.3 Dermal Exposure 2.2.3.1 Death No studies were located regarding lethality in humans or animals following dermal exposure to NDMA. 2.2.3.2 Systemic Effects No studies were located regarding systemic effects in humans following dermal exposure to NDMA. Limited information was located regarding systemic effects in animals following dermal exposure; no animal studies provided information on respiratory, cardiovascular, gastrointestinal, hematological, musculoskeletal, hepatic or renal effects. Dermal/Ocular Effects. Small ulcerations and scarring of the skin were observed in hairless mice that were treated once weekly with topical doses of 33.3 mg NDMA/kg for 20 weeks (Iversen 1980). No studies were located regarding NDMA-related ocular effects in animals. 39 2. HEALTH EFFECTS Other Systemic Effects. Barnes and Magee (1954) noted that daily application of 100 mg NDMA/kg to rats for 4 days had no effect on general condition. No studies were located regarding the following effects in humans or animals following dermal exposure to NDMA: 2.2.3.3 Immunological Effects 2.2.3.4 Neurological Effects 2.2.3.5 Developmental Effects 2.2.3.6 Reproductive Effects 2.2.3.7 Genotoxic Effects 2.2.3.8 Cancer No studies were located regarding carcinogenic effects in humans following dermal exposure to NDMA. A low incidence of lung adenomas (13%), but no skin tumors, developed in hairless mice that were treated once weekly with 33.3 mg/kg topical doses of NDMA for 20 weeks (Iversen 1980). Lung and skin tumors were not observed in historical control groups. Although Iversen (1980) concluded that the lung cancers were related to the topical applications of NDMA, it should be noted that the mice were housed 8 to a cage and could have licked the NDMA off each other or inhaled the compound due to its volatility. 2.3 RELEVANCE TO PUBLIC HEALIH Death. Oral LD50 values of 23 and 40 mg NDMA/kg have been reported for pregnant and nonpregnant rats, respectively (Nishie 1983, Druckrey 1967). Oral LDSOS have not been determined for NDMA in other species. LD5os for single doses of NDMA administered by intraperitoneal injection have also been reported; these values are consistent with the oral LD5os and include 26.5 mg/kg in rats (Barnes and Magee 1954), 42.7 mg/kg in rats (Heath 1962) and 19 mg/kg in mice (Friedman and Sanders 1976). Repeated oral exposure to NDMA resulted in decreased survival in rats, mice and all other species that have been tested. In general, doses ranging from approximately 0.1-5 mg/kg/day have produced death in animals after several days to several months of exposure. Variations in lethal doses appear to be attributable more to intraspecies differences than differences in frequency or method of oral treatment. With the exception of mink, there do not appear to be marked differences in sensitivity among the species that have been tested. Deaths resulting from a single exposure or repeated exposures for several days or several weeks are generally attributed to liver toxicity; deaths associated 40 2. HEALTH EFFECTS with longer duration (e.g., 20-40 weeks) exposures are due to liver tumor development. In general, lethal doses of NDMA and causes of death are similar among animal species. Human fatalities due to ingestion or inhalation of NDMA were also attributed to liver toxicity but adequate dose information is not available. Systemic Effects. Hepatotoxicity is the primary systemic effect of NDMA. Hepatotoxicity has been demonstrated in all animal species that have been tested, and has been observed in humans who were exposed to NDMA by ingestion or inhalation. The characteristic hemorrhagic necrosis caused by NDMA are particularly prevalent following exposure to acutely toxic single doses or repeated doses for short durations. Liver tumors are the predominant effect of longer duration exposures. The mechanism of NDMA— induced liver toxicity is not clearly understood but may be related to alkylation of cellular protein (Barnes and Magee 1954, Magee et a1. 1976, Diaz Gomez et al. 1981, 1983, Martino et a1. 1988). Although the hepatotoxicity of NDMA has been established unequivocally in numerous acute, intermediate and chronic duration oral studies with animals, relatively few of the studies delineate dose-response relationships and appropriate information regarding thresholds for this effect is not available. As noted for lethality, reported hepatotoxic doses for all species occur in the same general range with variations attributable more to intraspecies differences than treatment schedule or method. Human fatalities due to oral and inhalation exposure to NDMA have been reported in which hemorrhagic, necrotic and cirrhotic alterations in the liver were observed, indicating that NDMA produces similar hepatic effects in humans and animals. Therefore it is reasonable to expect that NDMA also will be hepatotoxic in humans at sublethal doses. Limited information is available regarding nonhepatic systemic effects of NDMA in humans. This information has been obtained from autopsies of victims accidentally exposed to NDMA vapors or poisoned after ingestion of NDMA. The effects can be described as a general bleeding tendency. Hemorrhages have been noticed in the gastrointestinal tract, heart, respiratory system and brain (Freund 1937; Kimbrough 1982). The mechanism by which NDMA could induce bleeding is not known, but the bleeding tendency could be a consequence of decreased formation of clotting factors resulting from liver damage, impairment of the clotting mechanism or decreased number or function of platelets. Jacobson et al. (1955), for example, showed that NDMA greatly increases prothrombin time in dogs exposed to NDMA by inhalation. It is also possible that hemorrhagic effects could be caused by effects of NDMA on tissues. Because of its irritant properties, it is not difficult to explain the occurrence of hemorrhages in tissues that have direct contact with NDMA (gastrointestinal bleeding after oral ingestion, or bleeding of the bronchi and trachea after inhalation). However, it remains unknown why gastrointestinal bleeding can occur following inhalation 41 2. HEALTH EFFECTS exposure. An alternative explanation could be that NDMA has a direct effect on the blood vessels. In fact, Ungar (1984, 1986) showed that oral treatment of hamsters with NDMA induced fragmentation of elastic fibers in portal vessels as well as denudation of the portal endothelium. Furthermore, autopsy of a victim of acute NDMA poisoning showed that "the central hepatic ‘ veins had lost their endothelial lining cells" (Kimbrough 1982). There is a relative paucity of information for nonhepatic systemic effects in animals because the emphasis of most studies was on hepatotoxicity or cancer, for which the liver is the primary target organ. Nonhepatic systemic effects that have been reported include gastrointestinal hemorrhage and congestion of several organs (kidney, lung, heart, spleen) in rats and/or mink, but the prevalence of these effects cannot be determined because these sites were examined infrequently. Immunological Effects. A single oral dose of NDMA near the oral LD50 did not reduce humoral immune response in rats, but a single intraperitoneal dose near the intraperitoneal LD50 reduced humoral immune response in mice (Waynforth and Magee 1974). A number of other recent studies have found that NDMA given by intraperitoneal injection alters humoral immunity and antibody-mediated host defense mechanisms (Kaminski et al. 1989; Thomas et al. 1985, Myers et a1. 1986, 1987, Scherf and Schmahl 1975, Holsapple et a1. 1983, 1984, 1985, Johnson et al. 1987a,b). Immunosuppression resulting from NDMA exposure is not believed to be a result of direct interaction between the reactive intermediaries of NDMA and splenic lymphocytes, thereby indicating a difference between the mechanisms of immunotoxicity and carcinogenicity/genotoxicity (Holsapple et a1. 1984). In vivo studies have shown that NDMA modulates the cellular immune response by altering the production and/or maturation/differentiation of bone marrow stem cells into functional macrophages (Myers et a1. 1986, 1987). In vitro tests identify the primary cell target of NDMA as the B-lymphocyte (Holsapple et al. 1984, 1985). Thus, it is likely that NDMA decreases the overall reactivity of both T- and B—lymphocytes. It is not known whether NDMA is likely to be immunosuppressive in humans. Developmental Effects. NDMA was fetotoxic to rats at oral doses that were toxic to the mother. Limited data indicate that these doses were not teratogenic for the rats. Oral administration of NDMA to mice resulted in increased perinatal deaths without histological abnormalities. It is not known whether NDMA could cause developmental effects in humans, but it could be a potential developmental toxicant at doses which are toxic to pregnant women. Reproductive Effects. Mice that were exposed to NDMA in drinking water prior to mating and during pregnancy and lactation showed an increase in the frequency of perinatal death among their offspring. Based on these data, NDMA could be considered a potential human reproductive toxicant. 42 2. HEALTH EFFECTS Genotoxic Effects. Several in vitro studies have examined genotoxic effects of NDMA in human cells. As indicated in Table 2-3, NDMA induced DNA repair and synthesis in human lymphoblasts, and sister chromatid exchange in human lymphocytes and fibroblasts. Genotoxicity of NDMA has been demonstrated consistently in numerous in vitro studies with non-human systems. As indicated in Table 2-3, NDMA was mutagenic in bacteria (Salmonella typhimurium, Escherichia coli), yeast (Saccharomyces cerevisiae), and mammalian cells (Chinese hamster V79 and ovary cells and mouse lymphoma L5178Y cells). NDMA induced unscheduled DNA synthesis and DNA repair and synthesis in rat, mouse and hamster hepatocytes. Treatment-related DNA fragmentation occurred in rat and human hepatocytes. Chromosomal aberrations occurred in Chinese hamster primary lung cells, rat ascites hepatoma cells, and rat esophageal tumor cells. Sister chromatid exchanges occurred in Chinese hamster ovary cells, Chinese hamster primary lung cells, human lymphoblasts and fibroblasts, and rat esophageal tumor and ascites hepatoma cells. In vivo studies (Table 2-4) have shown that NDMA methylates DNA, causes DNA fragmentation and induces DNA synthesis and repair in liver and other tissues of various species (e.g., rat, mouse, hamster, gerbil). NDMA induced chromosomal aberrations in hamster embryonic fibrolasts, sister chromatid exchanges in mouse bone marrow cells, and micronuclei in rat hepatocytes and rat and mouse bone marrow cells. The genotoxic effects indicated above occurred after inhalation, oral or intraperitoneal administration of NDMA. Sperm abnormalities were not seen in mice following intraperitoneal administration of NDMA. Sex linked recessive lethal mutations occurred in Drosophila melanogaster, which indicates potential heritable mutagenicity of NDMA. The weight of evidence indicates that NDMA is genotoxic in mammalian cells. In vitro studies with human cells, as well as in vitro and in vivo studies with animals and microbes, support this conclusion. Given the type and weight of genotoxicity evidence, it is appropriate to predict that NDMA poses a genotoxic threat to humans. Cancer. The oral carcinogenicity of NDMA has been demonstrated in numerous studies with various species of animals. Inhalation exposure to NDMA has been reported to be carcinogenic to rats and mice in two studies. The carcinogenicity of NDMA is also documented in numerous single and or weekly subcutaneous and intraperitoneal injection studies, and in studies in which NDMA was administered prenatally and to newborn animals. Many of the carcinogenicity studies of NDMA were conducted specifically to induce cancer for various purposes, such as investigations of structure-activity relationships and pathogenesis. Tumors in tissues other than the liver and respiratory system (e.g., kidney, testis) have not been observed often in many of the carcinogenicity studies; this appears to be attributable in part to limited examination of nonhepatic tissues. TABLE 2-3. Genotoxicity of N-Nitrosodimethylamine In Vitro Result Species With without Erxboint (Test System) Activation Activation References Gene mutation Salmonella tminuriun + + Araki et al. 1984, Bartsch et al. 1980, Langenbach et al. 1986, DeFlora et al. 1984, Prival and Mitchell 1981, Ishidate and Yoshikaua 1980 Escherichia oli + NT Araki et al. 1981., DeFlora et al. 1984 Saccharflces cerevisae + NT Jagannath et al. 1981, Frezza et al. 1983 Chinese Hamster V79 and + - Kuroki et al. 1977, Adair and Carver 1983, ovary cells O'Neill et al. 1982, Carver et al. 1981, Dickins et al. 1985, Bartsch et al. 1980, Katoh et al. 1982, Langenbach 1986, Hsie et al. 1978 Mouse lynphoma L5178Y cells + - Amacher and Paillet 1983, Clive et al. 1979 DNA fragmentation Rat hepatocytes NT + Bradley et al. 1982 Hunan hepatocytes NT + Martelli et al. 1985 Chromosomal Chinese hamster lung cells + NT Matsuoka et al. 1979, 1986, aberrations Ishidate and Yoshikaua 1980 Rat ascites hepatoma (AH668) and NT + lkeuchi and Sasaki 1981 rat esophageal (R1, R3) tunor cells '8 SlOHfidH HLTVHH EV TABLE 2-3 (continued) Result Species with Without Endpoint (Test System) Activation Activation References sister-chromatid Rat esophageal tunor, ascites hepatoma NT + Abe and Sasaki 1982, Ikeuchi and Sasaki 1981 exchange Human lymphocytes + - lnoue et al. 1983, Madle et al. 1987, Human fibroblasts + NT Tonkins et al. 1982 Chinese hamster ovary cells + NT/- Tomkins et al. 1982, Okinaka et al. 1981 Blazak et al. 1985 Chinese hamster V79 cells + - Madle et al. 1987, Sirianni and Huang 1987 Blazak et al. 1985 Chinese hamster primary lung cells + - Shimizu et al. 1984 DNA Danage Rat hepatocytes NT + Bermudez et al. 1982 DNA repair/ Rat hepatocytes + + Andrae and Schwarz 1981, synthesis Human lymphoblasts + NT Andrae et al. 1979 Mice hepatocytes NT + McQueen et al. 1983 Hamster hepatocytes NT + McQueen et al. 1983 Rat pancreatic cells NT - Steinmetz and Mirsalis 1984 NT = Not tested '3 SlDHJflH HL'IVHH 17‘7 TABLE 2-1.. Genotoxicity of N-Nitrosodimethylamine In Vivo Enqaoi nt Spec i es Result References (Test System) DNA methylation Rat, mouse, hamster + O'Connor et al. 1982, Banborschke et al. 1983, ard/or gerbil liver Pegg et al. 1981, Pegg and flui 1978, Stmpf et al. 1979 Hunan liver + Herron and Shank 1980 DNA fragmentation Rat liver and kidney elution + Brambilla et al. 1981, Petzold and Suenberg 1978, Abanobi et al. 1979, Beruudez et al. 1982 Mouse liver and kidney elution 4- Cesarone et al. 1982 DNA synthesis Fetal mouse kidney and liver + Bolognesi et al. 1988 and repair ‘ Mouse testes + Friednan and Stat) 1976, Cesarone et al. 1979 Rat liver + Bakke and Mirsalis 1984, Kornbrust and Diet: 1985, Doolittle et al. 1984 Rat respiratory cells + Doolittle et al. 1984 Rat spermatocytes - Doolittle et al. 1984 Sex-linked recessive lethal Droso ila melanogaster + Brodberg et al. 1987, Blount et al. 1985, nutations Lee et al. 1983 Sperm abnormalities Mouse - Hyrobek and Bruce 1975 Sister chromatid exchange Chinese hanster bone marrow +/- Neal and Probst 1983 Mouse bone narrow + Sharma et al. 1983, Bauknecht et al. 1977 chromosome aberrations Hamster enbryonic fibroblasts + Inui et al. 1979 Micronucleus Rat bone narrow +/- Trzos et al. 1978 Rat hepatocytes + Mehta et al. 1987, Tates et al. 1980 Mouse bone marrow + Odagiri et al. 1986, Bauknecht et al. 1977 Hamster enbryonic fibroblasts Inui et al. 1983 'Z SIDHJJH HLTVEH SW 46 2. HEALTH EFFECTS There is increasing evidence, derived from in vitro and in vivo metabolic studies, indicating that the carcinogenic effects of NDMA are due to a metabolite rather than the compound itself (Singer 1979). NDMA is converted into an alkylating (methylating) agent after metabolism by microsomal mixed—function oxidases. This process occurs principally in the liver and to a lesser extent in kidney and lungs, and results in the methylation of cellular macromolecules such as DNA, RNA and other proteins. Methylation occurs at several positions in DNA including N-l, N-3 or N-7 of deoxyadenosine; N-3, N-7 or O6 of deoxyguanosine; N-3 of deoxycytidine; and O2 or O of thymidine. Experimental evidence indicates that methylation at the 06-position of guanine may be responsible for the carcinogenic activity of nitrosamines in general, however, the carcinogenic potential of other methylated products cannot be ruled out. The methylation of DNA by NDMA has been studied extensively (e.g., Bamborschke et a1. 1983, O’Connor et a1. 1982, Pegg et a1. 1981, Pegg and Hui 1978, Stumpf et al. 1979.). The carcinogenic properties of NDMA, and nitrosamines in general, have been extensively studied. It is of considerable interest that, despite its ubiquitous distribution, NDMA induces tumors in a limited number of organs and tissues and that there are marked differences in this response among animal species. Differences in pharmacokinetics properties seem to play an important role in the carcinogenic action of NDMA (Pegg 1980, Lijinsky 1987). For example, the degree of hepatic extraction from the portal blood seems to determine whether tumors develop in extrahepatic sites. Therefore, large doses of NDMA tend to induce extrahepatic tumors (spill—over effect). In addition, metabolic activating systems and repair mechanisms may not operate at the same rates in different organs and different species. Route of administration also seems to be a factor in NDMA carcinogenesis since different responses are seen in a particular species when different routes of exposure are used. This suggests that rates of absorption can determine the site of tumor development. Based on the unequivocal evidence of carcinogenicity in animals, it is reasonable to anticipate that NDMA will be carcinogenic in humans. It is important to recognize that this evidence also indicates that oral exposures of acute and intermediate duration are sufficient to induce cancer. 2.4 LEVELS IN HUMAN TISSUES AND FLUIDS ASSOCIATED WITH HEALTH EFFECTS A considerable amount of DNA methylation in the liver of a suspected NDMA poisoning case was reported by Herron and Shank (1980). Based on studies in rats, in which the amount of DNA alkylation could be correlated with known amount of orally administered NDMA, the authors estimated that the victim had been exposed to a dose of 20 mg/kg or more of NDMA. No other studies were located regarding levels of NDMA or its metabolites in human tissues and fluids associated with effects. Several analytical methods have been developed to determine levels of NDMA in human tissues and fluids. These methods are described in Chapter 6. 47 2. HEALTH EFFECTS 2.5 LEVELS IN THE ENVIRONMENT ASSOCIATED WITH LEVELS IN HUMAN TISSUES AND/OR HEALTH EFFECTS Although data relating specific amounts of NDMA in the environment with levels in human tissues and/or health effects have not been reported, some qualitative information is available. This information, given below, has to be interpreted with caution since results were not always rigorously reported or evaluated, and endogenous formation of NDMA was not quantitated. A high incidence of nasopharyngeal carcinoma was found in Tunisia (North Africa), Southern China and Greenland among populations who consume foods with a high content of volatile nitrosamines (Poirier et a1. 1987). Lu et a1. (1987) found a positive correlation between the amount of NDMA and other nitrosamines in the gastric juice of Chinese with a high incidence of esophageal carcinoma. Wild et al. (1987) observed a positive relationship between levels of 06—methyldeoxyguanosine (implicated in the initiating process of nitrosamine—induced cancer) and incidences of esophageal and stomach cancer in the province of Linaxian, China. Yu and Henderson (1987) reported finding a high incidence of nasopharyngeal carcinoma in individuals from Hong Kong, who are known to consume from early in life considerable amounts of Cantonese—style salted fish, which has a high content of NDMA and other nitrosamines. 2.6 TOXICOKINETICS 2.6.1 Absorption 2.6.1.1 Inhalation Exposure No studies were located regarding the rate and extent of absorption of NDMA following inhalation exposure of humans or animals to NDMA. However, it can be inferred that NDMA is absorbed from the air since it can be detected in the urine of rats (Klein and Schmezer 1984) and dogs (Raabe 1986) after inhalation exposure. Absorption is also indicated by reports of human deaths following inhalation of NDMA (see Section 2.2.1.1). 2.6.1.2 Oral Exposure No studies were located regarding the absorption of NDMA following oral exposure of humans. The absorption of NDMA from the gastrointestinal tract of animals is fast. Less than 2% of the labelled compound could be recovered from the gastrointestinal tract 15 minutes after oral administration of 14C-NDMA to rats (Diaz Gomez et al. 1977). Absorption seems to be independent of the dose administered (Diaz Gomez et a1. 1977). In the rat, NDMA is absorbed much faster from the small intestine than from the stomach, in isolated preparations (Heading et a1. 1974) and in vivo (Pegg and Perry 1981). 48 2. HEALTH EFFECTS Ishiwata et al. (1977) reported that the disappearance curve of NDMA from isolated guinea pig stomach and small intestine follows first order kinetics. 2.6.1.3 Dermal Exposure No studies were located regarding the absorption of NDMA following dermal exposure of humans. Indirect evidence indicating that NDMA may be absorbed through the skin was found in a study published by Iversen (1980) in which topical application of NDMA induced lung adenomas in mice. The results from Iversen, however, should be interpreted with caution since the mice were housed 8 to a cage and could have licked the NDMA from each other and also could have inhaled this volatile compound. 2.6.2 Distribution Unmetabolized NDMA was found to be evenly distributed among the main organs of mice and rats shortly after i.v. injection to animals in which the metabolism of NDMA had been inhibited (Magee 1956; Johansson and Tjalve 1978). Wishnok et al. (1978) reported a similar finding in rats following i.p. injections. Johnson et al. (1987a) reported that one hour after a dose of 6 mg C-NDMA/kg was administered by intraperitoneal injection to mice, the liver contained two times as much radioactivity as the kidney, spleen and thymus. 2.6.2.1 Inhalation Exposure No studies were located regarding the distribution of NDMA following inhalation exposure of humans or animals. 2.6.2.2 Oral Exposure No studies were located regarding the distribution of NDMA following oral exposure of humans. Daugherty and Clapp (1976) reported that 15 minutes after oral administration of C-NDMA to mice, the relative amounts of radioactivity in the homogenates of heart, forestomach, esophagus, liver and lung were 1, 2, 3, 10 and 70, respectively. The differences could be attributed to different tissue affinity, transport and/or metabolism. Measurable amounts of NDMA were reported in blood, liver, kidney, lungs and brain of mice exposed to 5 mg NDMA/kg/day in drinking water for up to 4 weeks (Anderson et al. 1986). NDMA has been detected in maternal blood, placenta, fetus and amniotic fluid of pregnant Syrian hamsters for up to 2 hours after a single subcutaneous dose of 12.5 mg/kg of the chemical (Althoff et al. 1977). 49 2. HEALTH EFFECTS Liver and kidney DNA from lA-day-old rats became labelled after treating the nursing mothers with 14C-NDMA by gavage (Diaz Gomez et a1. 1986). 2.6.2.3 Dermal Exposure No studies were located regarding the distribution of NDMA following dermal exposure of humans. The study by Iversen (1980), in which lung adenomas were noticed in mice after skin application of NDMA, indicates that this chemical (or a metabolite) was distributed to the lungs. 2.6.3 Metabolism Evidence from in vitro and in vivo studies with rodents indicates that NDMA is metabolized by hydroxylation of the a-carbon, followed by formation of formaldehyde, molecular nitrogen and a methylating agent, which is considered to be the carcinogenic form (Lotlikar et a1. 1975; Czygan et a1. 1973). Recent evidence suggests that a significant proportion of NDMA is metabolized via a denitrosation mechanism. The latter mechanism takes place in rats in vivo, as indicated by the urinary excretion of labelled methylamine after i.v. administration of 14C—NDMA (Keefer et a1. 1987), and in human liver microsomes (Yoo et a1. 1988). The metabolism of NDMA is summarized in Figure 2-3. Metabolism of NDMA varies among species (Prassana et a1. 1985; Montesano et a1. 1982). Age of the animal and route of administration can also influence the rate of metabolism of NDMA (Phillips et a1. 1975). In addition, at varying doses, different forms of enzymes appear to be responsible for NDMA metabolism (Kroeger-Koepke and Michejda 1979; Lotlikar et al. 1978). 2.6.3.1 Inhalation Exposure No studies were located regarding the metabolism of NDMA following inhalation exposure of humans or animals. 2.6.3.2 Oral Exposure No studies were located regarding the metabolism of NDMA following oral exposure of humans. Phillips et al. (1975) demonstrated that NDMA is metabolized at a lower rate when given orally to rats than when administered by parenteral routes. 50 2. HEALTH EFFECTS NO I OHS—N-CHJ donllrosolion N-niivosodimelhylomino H H 9450 l ' | a-hydroxylaHon iso.nxym. H-N-CH3 + N02- + (H‘C=0) molhylomint nilriio (olmoldonydn (puslulolod) NO I HO‘CH2-N-CH3 a-nydroxy N-nilvcsamlno //// N0 1 I Coulomotlzeiion H-CHO + H-N—CH3 ——————~——» CHs-NnN-OH dloxohydrolldo H + CHs—NEN + OH' olkyldluzonium Ion (molnylnllng agent for substrate 1) ng CH3X + N2 nitrogen formaldghyd. N-nllrosomolnylomlno Figure 2-3. Metabolism of N—Nitrosodimethylamine Source: Crygan et a1. 1973; Keefer et a1., 1987; Lotikar et a1. 1975; Ybo et a1. 1988 51 2. HEALTH EFFECTS 2.6.3.3 Dermal Exposure No studies were located regarding the metabolism of NDMA following dermal exposure of humans or animals. 2.6.4 Excretion Labelled C02 can be detected in the exhaled air 1 hour after i.p. administration of 14c-NDMA to rats (Phillips et a1. 1975). Hemminki (1982) administered labelled NDMA by intraperitoneal injection to rats and was able to detect three main radioactive fractions in the urine over a period of 5 days. Fraction I was composed of radioactive aminoacids, fraction II of allantoin and a metabolite of thiazolidine-4-carboxylic acid, and fraction III of 7-methy1guanine. 2.6.4.1 Inhalation Exposure Klein and Schmezer (1984) reported that 10-30% of NDMA is excreted by exhalation after exposing rats to the chemical during 10 minutes by endotracheal intubation. In beagle dogs, 23% of the administered radioactive label is exhaled in 30 minutes after a 3 hour inhalation exposure (Raabe 1986). 2.6.4.2 Oral Exposure Spiegelhalder et a1. (1982) reported that, in a 24 hour period, human volunteers excreted in the urine between 0.5 and 2.4% of an ingested dose of 12-30 pg of NDMA added to drinking fluids containing ethanol. Unchanged NDMA was recovered in the urine and feces of rats up to 24 hours after a single oral dose of 50 mg (Magee 1956). Swann et al. (1984) did not detect labelled NDMA in the urine of rats after oral administration of 30 pg/Kg of 14C-NDMA in water. Phillips et a1. (1975 determined that after administration of a single oral dose of 5 mg of 1 C-NDMA to female rats the maximum rate of 14C02 production was 12.4% of the dose/hour, and that 48% of the dose could be recovered as 14C02 in the exhaled air in 7 hours and 5.7% as 14C (total label) in a 24 hour urine sample. 2.6.4.3 Dermal Exposure No studies were located regarding the excretion of NDMA following dermal exposure of humans or animals. 2.7 INTERACTIONS WITH OTHER CHEMICALS NDMA is normally formed by bacteria in the human stomach and small intestine, but not in the large intestine (Archer et al 1982; Spiegelhalder and Preussmann 1985; Zeisel et a1. 1988). Also, rats and guinea pigs have 52 2. HEALTH EFFECTS been shown to make NDMA in their stomachs (Hashimoto et a1. 1976; Omori et a1. 1979). Small amounts of NDMA are formed in the saliva of humans; concentrations can vary from 4 to 10 pg/mL depending on pH and type of food in the mouth (Rao et al. 1982). NDMA formation in the saliva can be increased by chemicals such as chlorogenic acid, which is found in coffee, and decreased by a number of synthetic additives, as well as caffeic acid, tannic acid and ascorbic acid, which are found in coffee, tea, and citrus fruits, respectively. Consumption of alcohol has been shown to have complicated effects on the toxicity of NDMA. Rats that received alcohol (ethanol or isopropanol) by gavage for 2 days before receiving NDMA had more liver damage with the alcohol than without it (Lorr et a1. 1984; Maling et a1. 1975). Increased levels of plasma glutamic pyruvate transaminase were monitored and used as a sign of liver damage. Another study showed that 4 weeks of ethanol pretreatment in rats worsened the effects on DNA repair that occurred following DNA alkylation induced by NDMA (Mufti et a1. 1988). There is at least one other study in rats, however, that showed that 23 days of pretreatment with ethanol decreased the hepatotoxicity of NDMA (Gellert et a1. 1980). Other substances to which people are exposed have been shown to alter the toxic effects of NDMA in rats. Vitamin E and calcium channel blocking agents have been shown to decrease the hepatotoxicity associated with NDMA (Landon et a1. 1986; Skaare and Nafstad 1978). Selenium increased the toxic effect of NDMA on the liver (Skaare and Nafstad 1978) and cadmium increased the carcinogenic effect of NDMA in the kidney (Wade et a1. 1987). NDMA induced higher incidences of stomach cancer in rats fed diets low in zinc than in those fed normal diets (Ng et a1. 1984). Rats fed diets low in copper developed more kidney tumors from NDMA than rats fed normal diets (Carlton and Price 1973). In contrast, rats given NDMA and cupric acetate had fewer tumors than rats given NDMA (Yamane et a1. 1984). Although these data indicate that simultaneous administration of other chemicals may augment NDMA toxicity in animals, it not clear how these simultaneous exposures may occur in humans. 2.8 POPULATIONS THAT ARE UNUSUALLY SUSCEPTIBLE People with chronic renal failure produce more NDMA in their small intestines due to increased levels of bacterial growth than normal people do (Lele et al. 1983). This increase in NDMA can be blocked by injections of ascorbic acid or antibiotics, but is potentiated by alcohol (Lele et a1. 1987). People who consume alcohol may be unusually susceptible to NDMA for reasons discussed in Section 2.7. 2.9 ADEQUACY OF THE DATABASE Section 104 (i) (5) of CERCLA, directs the Administrator of ATSDR (in consultation with the Administrator of EPA and agencies and programs of the 53 2. HEALTH EFFECTS Public Health Service) to assess whether adequate information on the health effects of NDMA is available. Where adequate information is not available, ATSDR, in cooperation with the National Toxicology Program (NTP), is required to assure the initiation of a program of research designed to determine these health effects (and techniques for developing methods to determine such health effects). The following discussion highlights the availability, or absence, of exposure and toxicity information applicable to human health assessment. A statement of the relevance of identified data needs is also included. In a separate effort, ATSDR, in collaboration with NTP and EPA, will prioritize data needs across chemicals that have been profiled. 2.9.1 Existing Information on Health Effects of N-Nitrosodimethylamine Information regarding health effects of NDMA in humans is limited to case reports of fatalities due to hepatotoxicity following ingestion or inhalation. Health effects of NDMA in animals have been investigated in numerous oral studies and several inhalation and dermal studies. As indicated in Figure 2—4, animal oral data are available for lethality, systemic toxicity, immunological effects, neurological effects, developmental effects, reproductive effects, genotoxic effects and cancer. These data indicate that hepatotoxicity and cancer are the most prominent NDMA-related effects. 2.9.2 Data Needs Single Dose Exposure. Information on lethality in rats following single oral doses, including two LD50 values, are available. Information on hepatic effects in rats due to single oral exposures are also available. Additional single dose oral studies with rats would provide more information on thresholds for lethality and hepatotoxicity, and on nonhepatic effects. Studies on species other than the rat would provide data on interspecies differences. Single-exposure inhalation experiments provide limited information on lethality in rats, mice and dogs, and dermal/ocular effects in rats; additional studies could corroborate these data as well as provide NOAELs. Single application dermal studies would provide information on lethality and skin and eye irritation. Repeated Dose Exposure. Numerous repeated dose studies of intermediate duration have been conducted with rats, mice and other species. These studies provide extensive information on doses and treatment schedules that are lethal and hepatotoxic but do not adequately identify thresholds for these effects, particularly in species that may be more sensitive (e.g., mink). Additional repeated dose oral studies designed to examine tissues other than the liver could provide useful information on nonhepatic systemic effects of NDMA. Oral studies conducted over periods longer than 20—30 weeks may not be necessary as sufficient evidence indicates that cancer will be the predominant effect. Repeated exposure inhalation studies could provide 54 HEALTH EFFECTS 2. Inhalation Oral Dermal HUMAN Inhalation . Oral Dermal ANIMAL . Existing Studies Existing Information on Health Effects of FIGURE 2—4. N-Nitrosodimethylamine 55 2. HEALTH EFFECTS information on concentrations associated with lethality and systemic effects. Chronic Exposure and Carcinogenicity. Chronic oral and inhalation studies of NDMA have been conducted with rats and mice. These studies indicate that the predominant effect of chronic exposure to NDMA is cancer. As low doses of NDMA have been tested in chronic oral studies and it is established that intermediate duration exposure to NDMA is sufficient to induce cancer, additional chronic studies may not be needed. Genotoxicity. The genotoxic potential of NDMA is established unequivocally. Only several studies, however, evaluated genotoxic effects in animals following oral or inhalation exposure to NDMA, and only several in vitro studies evaluated human cells. Additional studies, particularly assays with human cells and assays providing information on the potential for heritable mutations, would add to the data base on genotoxicity. Reproductive Toxicity. Oral exposure to NDMA for 75 days prior to mating had no significant effect on time-to-conception in mice. Other reproductive indices or species have not been evaluated. Histological examinations of reproductive organs of animals exposed in subchronic and chronic studies would provide relevant data. Multigenerational or continuous breeding studies would provide further information regarding reproductive effects of NDMA in animals, which may be related to possible reproductive effects in human. Developmental Toxicity. Evidence indicates that NDMA is fetotoxic to rats and mice, but NOAELs have not been defined. Well-conducted developmental studies using several exposure levels and environmentally relevant routes of exposure could provide the dose-response information necessary to determine the threshold for fetotoxicity and to determine the possible relevance and risk for humans. Additional studies also could determine if NDMA is a transplacental carcinogen. Immunotoxicity. Information regarding immunological effects of NDMA in humans is not available. Immunosuppression by NDMA has been demonstrated in a number of intraperitoneal injection studies, but not in an oral study, with mice. Specific immunotoxicity tests or a battery of immunotoxicity tests in which NDMA is administered by the oral route would provide a better assessment of possible immunotoxic effects. Sensitization tests in animals could provide information on whether an allergic response to NDMA is likely in humans. Additional studies also could determine if NDMA is a transplacental carcinogen. Neurotoxicity. Dogs that were orally treated with NDMA reportedly experienced central nervous system depression, but it is likely that this effect is a consequence of liver damage rather than direct neurotoxicity. Additional information pertaining to neurotoxicity was not found. 56 2. HEALTH EFFECTS Neurotoxicity tests in animals exposed to NDMA could provide additional information on possible neurotoxic effects. Epidemiological and Human Dosimetry Studies. The only information available concerning effects of NDMA in humans comes from cases of acute poisoning and subsequent death. In these cases, hemorrhagic and necrotic alterations and cirrhosis of the liver were observed. On the other hand, effects in animals have been well documented (Section 2.2). Attempts have been made to measure occupational exposure to NDMA, in particular in the rubber industry. Unfortunately these attempts have failed because NDMA is metabolized almost completely to 002 and water. Excretion rates for NDMA measured in experimental animals are in the order of 0.02% of the ingested dose (Spiegelhalder 1984). Although unchanged NDMA is unlikely to be detected in the urine, it may be possible to measure urinary excretion of nonspecific DNA adducts (e.g., 7-methylguanine). As stated by Spiegelhalder (1984), limited information is available on airborne exposures of individual workers for the following reasons: 1) usually workers are exposed to a variety of chemicals and there is cross-contamination between jobs, 2) transfers from job to job involve different exposures, 3) increases in cancer incidences most likely result from exposures that occurred in the past, when no exposure data were available, and 4) no comprehensive study has been conducted so far. Epidemiology studies of individuals who live in areas where NDMA has been detected are necessary to obtain information on whether NDMA induces effects in humans similar to those seen in animals. Biomarkers of Disease. Since acute NDMA poisoning in humans caused severe liver disease, sensitive clinical biochemistry liver function tests might detect early hepatic damage from toxic exposure to NDMA. Recently, Wild et al. (1987), using a radioimmunoassay, were able to detect elevated levels of the promutagenic lesion 06-methyldeoxyguanosine in DNA of esophageal cells from individuals with high incidence of esophageal and stomach cancer. These individuals were found to consume foods with a relatively high content of nitrosamines. Disease Registries. The only known health effects of NDMA on humans are those obtained from acute poisoning cases, in which postmortem examination revealed severe liver damage. If disease states attributed to exposure to NDMA could be identified by epidemiological studies, the number of individuals affected, the exposure levels involved, and the factors associated with identifying the disease in a given population, such as, the vicinity to hazardous waste sites or industrial plants, could be determined. Bioavailability from Environmental Media. No studies were located regarding the bioavailability of NDMA from environmental media. Since NDMA has been detected in ambient air, water and soil (ppb levels), it is important to determine if NDMA can be absorbed by humans from environmental samples. It must be noted that NDMA has been found in trace amounts in some foods and beverages and that endogenous formation of NDMA has been found to occur from the nitrosation of amines in the gastrointestinal tract. An 57 2. HEALTH EFFECTS understanding of the bioavailability of NDMA from environmental media may be obtained by studying the biological fluids of individuals exposed in the workplace or through the ingestion of NDMA-containing foods and beverages. The limited information available regarding absorption parameters of NDMA in experimental animals indicates that NDMA is rapidly absorbed from the gastrointestinal tract; therefore, one can assume that if water or soil contaminated with NDMA are ingested, NDMA will be readily absorbed. _ Food Chain Bioaccumulation. No studies were available concerning food chain bioaccumulation of NDMA from environmental sources. NDMA has been detected in samples of cooked fish and meat. However, occurrence of NDMA in these samples is not the result of bioaccumulation but is the result of formation during preservation and/or cooking (Scanlan 1983). Estimation techniques have been used to determine that NDMA would not bioaccumulate in lipids of fish (see Section 5.3.1). Based on this limited amount of information, it is speculated that human exposure to NDMA through diet is not the result of food chain bioaccumulation. Monitoring for the accumulation of NDMA in organisms from several trophic levels could be used to support this conclusion. Absorption, Distribution, Metabolism, Excretion. Examination of Section 2.6 clearly indicates that oral administration of NDMA has been the preferred route for studying its absorption, distribution, metabolism and excretion. This is not surprising since oral administration is easier to monitor when compared to other routes. The oral route seems to be the most pertinent to study since humans are most likely to be exposed to nitrosamines orally. Toxicokinetic data with regard to dermal and inhalation exposure of NDMA are clearly lacking. Furthermore, dermal and inhalation exposures may lead to different metabolic pathways and patterns of distribution and excretion, which could account for differences in the degree of toxicity exhibited by different routes of exposure. The metabolism of NDMA in isolated microsomal preparations seems to be well understood, but studies with cultured human cells could provide additional useful information. However, exploration of the denitrosation mechanism as an alternative to a-hydroxylation requires more attention. Determination of the urinary excretion of NDMA in control human volunteers and in individuals known to consume foods with high contents of nitrosamines could provide information concerning absorption and excretion of the xenobiotic. Comparative Toxicokinetics. No studies were located regarding comparative toxicokinetics of NDMA in vivo. In vivo studies are available indicating differences in hepatic 06-methylguanine repair activity among rodent species (O'Connor et a1. 1982). A report by Prasanna et al. (1985) indicates that the in vitro metabolism of NDMA by liver microsomes from hamsters, rats and chickens is qualitatively similar, but with different rates. Montesano et al. (1982) showed that liver slices from humans have a metabolic capacity to activate NDMA similar to that found in rats and slightly lower than that found in liver slices from hamsters. Differences among species in the toxic responses to a chemical can be attributed to 58 2. HEALTH EFFECTS differences in the toxicokinetic parameters. This seems to be particularly true for N-nitrosamines in general (Lijinsky 1987). The fact that a number of factors (animal species, route of exposure, dosing schedule) appear to determine the organ-specificity and the severity of the effect of NDMA indicates that caution must be exercised when assuming possible effects in humans. Although little information is available regarding the toxicokinetics of NDMA in humans, analysis of NDMA in the urine of individuals accidentally exposed to the chemical or of individuals consuming foods with a relatively high content of NDMA could provide quantitative information on absorption and excretion. 2.9.3 On-going Studies Two studies regarding the immunotoxicity of NDMA are known to be on- going (Federal Research In Progress, 1988). One is investigating the immunosuppressive activity of subchronic and chronic administration of NDMA, specifically the in vitro antibody response of NDMA treated spleen cell suspensions to a number of mutagens. This research is being performed by Holsapple at Virginia Commonwealth University. A second study, performed by Schook at the University of Illinois, is attempting to identify molecular mechanisms for the immunosuppressive effects of NDMA. In research being conducted by Anderson at the Division of Cancer Etiology, National Cancer Institute, NDMA is being examined for its ability to cause neurogenic tumors in mice by transplacental exposure. In studies sponsored by NIEHS, Faustman at the Univerity of Washington is evaluating NDMA and other related N-nitroso compounds for their in vitro developmental toxicity (Faustman 1989). A number of ongoing studies are investigating the metabolism of NDMA (Federal Research in Progress, 1988). These include N-nitroso compound detoxification by Jensen at Temple University, Philadelphia, PA, formation and metabolism of nitrosamines in pigs by Magee at Temple University, metabolism and genotoxicity of nitrosamines in rats by Rogan at the University of Nebraska, Omaha, NE, and enzymology of nitrosamine metabolism in rats, mice and rabbits in a NCI-sponsored study by Yang at the University of Medicine and Dentistry, Newark, NJ. Other studies sponsored by NCI are being conducted to find means of shifting the balance of the metabolic pathway towards increasing inactivation and characterizing the possible role of a-nitrosamino radicals in the metabolism of NDMA (written communication, Keefer 1989). 3.1 3-1. 3.2 3-2. 59 3. CHEMICAL AND PHYSICAL INFORMATION CHEMICAL IDENTITY Data pertaining to the chemical identity of NDMA are listed in Table PHYSICAL AND CHEMICAL PROPERTIES The physical and chemical properties of NDMA are presented in Table 60 3. CHEMICAL AND PHYSICAL INFORMATION TABLE 3-1. Chemical Identity of N-Nitrosodimethylamine Value Reference Chemical name Methanamine, N-methyl-N-nitroso CAS 1988 Synonyms N-nitrosodimethylamine; CAS 1988 dimethylnitrosamine; DMNA; DMN; NDMA Trade name(s) ND Chemical Formula CZ H6 N2 0 SANSS 1988 Chemical Structure (CH3)2N-N=O SANSS 1988 Identification Numbers: CAS Registry 62-75-9 CAS 1988 NIOSH RTECS IQ0525000 RTECS 1988 EPA Hazardous Waste P082 RTECS 1988 OHM-TADS 7217418 OHM-TADS 1988 DOT/UN/NA/IMCO ND HSBD 1667 HSDB 1988 NCI . ND ND = No Data CAS = Chemical Abstract Service NIOSH = National Institute for Occupational Safety and Health RTECS = Registry of Toxic Effects of Chemical Substances EPA = Environmental Protection Agency OHM-TADS a Oil and Hazardous Materials - Technical Assistance Data Base DOT/UN/NA/IMCO = Department of Transportation/United Nations/North America/International Maritime Consultive Organization HSDB = Hazardous Substances Data Bank NCI = National Cancer Institute 61 3. CHEMICAL AND PHYSICAL INFORMATION TABLE 3-2. Physical and Chemical Properties of N-Nitrosodimethylamine Property Value Reference Molecular weight 74.08 Weast 1983 Color yellow IARC 1978 Physical State liquid IARC 1978 Melting point ~25°C (estimated) Lyman 1985 -50°C (estimated) EPA 1986 Boiling point 154°C Weast 1983 Specific gravity, 20/4°C Weast 1983 Odor Odor threshold Solubility Water Organic solvents Partition coefficient Log octanol/water Log Koc Vapor pressure Henry’s Law constant at 37°C at 20°C 1.0059 No distinct odor Not available Miscible Soluble in alcohol, ether, other organic solvents -0.57 1.07 (estimated using Equation 4-8) 2.7 mm Hg (20°C) 1.99x10'6 atm-m3/mol 2.63x10'7 atm-m3/mol (estimated using vapor pressure and water solubility data) Frank and Berry 1981 Mirvish et al. 1976 Weast 1983, IARC 1978 Hansch and Leo 1985 Lyman 1982 Klein 1982 Mirvish et a1. 1976 62 3. CHEMICAL AND PHYSICAL INFORMATION TABLE 3—2 (continued) Property Value Reference Autoignition temperature, °C ND Flashpoint, open cup ND Flammability limits in air ND Conversion factors ppm (v/v) to mg/m3 in air (20°C) ppm (v/V) x 3.08 = mg/m3 mg/m3 to ppm (v/v) in air (20°C) mg/m3 x 0.325 = ppm (V/v) ND = no data 63 4. PRODUCTION, IMPORT, USE, AND DISPOSAL 4.1 PRODUCTION NDMA is not produced for commercial use in the United States (HSDB 1988). The public portion of the U.S. EPA TSCA Production File indicates that during 1977, the Ames Laboratories in Milford, CT and Columbia Organics in Columbia, SC both prepared small research quantities of this chemical. Eastman-Kodak in Rochester, NY and Teledyne McCormick Selph, an importer, supplied no NDMA during 1977, although both had the capability to produce/import this compound and had done so in the past (EPA 1977). Small research quantities of this chemical presently are available from Sigma Chemical Co. and Aldrich Chemical Co. NDMA can be prepared by reaction of nitrous acid with dimethylamine or by addition of acetic acid and sodium nitrite to dimethylamine (HSDB 1988). 4.2 IMPORT Data pertaining to the import of NDMA into the U.S. were not located in the available literature. 4.3 USE NDMA is prepared in laboratory-scale quantities solely for use as a research chemical (HSDB 1988). NDMA was formerly used (prior to April 1, 1976) as an intermediate in the production of 1,1-dimethylhydrazine, a storable liquid rocket fuel, which was believed to have contained up to 0.1% NDMA as an impurity (IARC 1978). NDMA has also been used or has been proposed for use as an antioxidant, additive for lubricants, and as a softener for copolymers (Windholz 1983). NDMA has also been used as a solvent and rubber accelerator (Hawley 1981). 4.4 DISPOSAL Combustion in an incinerator equipped with an afterburner and NOx scrubber is the recommended method for disposing NDMA. Liquid wastes should be neutralized, if necessary, filtered to remove solids, and then put into closed polyethylene containers for transport. All equipment should be thoroughly rinsed with solvent, which should be added to the liquid waste for incineration. Great care should be practiced to insure that there is no contamination on the outside of the solvent container. If possible, solid waste should also be incinerated. If this is not possible, the nitrosamine should be extracted from the waste and the extract should be handled as a liquid waste. Any rags, papers or other materials which are contaminated during the disposal process should be incinerated. Contaminated solid materials should be enclosed in sealed plastic bags that are labeled cancer- suspect agent, with the name and amount of carcinogen. Bags should be stored in well-ventilated areas until they are incinerated (HSDB 1988). Nitrosamine residues generated in laboratory research or accidental spills 64 4. PRODUCTION, IMPORT, USE, AND DISPOSAL in research laboratories should be diluted to a concentration of less than 10 pg/L and then reduced to innocuous amines, ammonia, or alcohols by aluminum-nickel alloy powder and aqueous alkali. This method of disposal is applicable to a variety of media (water, mineral oil, olive oil, dimethylsulfoxide, solutions of agar gel), but is not recommended for use in solutions of acetone or dichloromethane because reactions are slow and incomplete. After the reduced reaction mixture is filtered, the liquid can be disposed of by pouring it over a sufficient amount of absorbent material to convert it to a solid waste for incineration. The filtercake is discarded with non-burnable solid wastes (HSDB 1988). Other methods of destruction of NDMA in laboratory wastes (e.g., using hydrobromic acid or potassium permanganate/sulfuric acid) are described by IARC (1982). 4.5 ADEQUACY OF THE DATA BASE Section 104 (i) (5) of CERCLA, directs the Administrator of ATSDR (in consultation with the Administrator of EPA and agencies and programs of the Public Health Service) to assess whether adequate information on the health effects of NDMA is available. Where adequate information is not available, ATSDR, in cooperation with the National Toxicology Program (NTP), is required to assure the initiation of a program of research designed to determine these health effects (and techniques for developing methods to determine such health effects). The following discussion highlights the availability, or absence, of exposure and toxicity information applicable to human health assessment. A statement of the relevance of identified data needs is also included. In a separate effort, ATSDR, in collaboration with NTP and EPA, will prioritize data needs across chemicals that have been profiled. 4.5.1 Data Needs Production, Use, Release, and Disposal. Uses, methods of synthesis, and methods of disposal for NDMA are described in the literature and there does not appear to be a need for further information on these topics. Lack of information pertaining to the import of this compound is not surprising since this compound has no commercial applications. Data regarding the amount of NDMA released to air, water, and soil would be useful in order to establish potential sources of exposure and levels of exposure from environmental media. In particular, information releases from hazardous waste landfills and industries in which this compound is inadvertently formed may help determine whether people living in the vicinity of these sites are exposed to elevated levels of this compound. According to the Emergency Planning and Community Right to Know Act of 1986 (EPCRTKA), (§313), (Pub. L. 99-499, Title III, §3l3), industries are required to submit release information to the EPA. The Toxic Release Inventory (TRI), which contains release information for 1987, became available in May of 1989. This database will be updated yearly and should provide a more reliable estimate of industrial production and emission. 65 5. POTENTIAL FOR HUMAN EXPOSURE 5.1 OVERVIEW NDMA is not an industrially or commercially important chemical; nevertheless, it can be released into the environment from a wide variety of manmade sources. This is due to the inadvertent formation of NDMA in industrial situations when alkylamines, mainly dimethylamine and trimethylamine, come in contact and react with nitrogen oxides, nitrous acid, or nitrite salts, or when trans-nitrosation via nitro or nitroso compounds occurs. Thus, potential exists for release into the environment from industries such as tanneries, pesticide manufacturing plants, rubber and tire manufacturers, alkylamine manufacture/use sites, fish processing industries, foundries and dye manufacturers. At this time, NDMA has been found in at least 1 out of 1177 hazardous waste sites on the National Priorities List (NFL) in the United States (VIEW Database 1989). Nitrosation reaction may also result in the formation of NDMA in the environment. In air, NDMA may form as a product of the nighttime reaction of dimethylamine with NOx. In water and soil, NDMA forms by the reaction of widely-occurring primary, secondary or tertiary amines in the presence of nitrite. In the ambient atmosphere, NDMA should be rapidly degraded upon exposure to sunlight. The half-life for direct photolysis of NDMA vapor is on the order of 5 to 30 minutes. In surface water exposed to sunlight, NDMA would also be subject to photolysis. 0n soil surfaces, NDMA would be subject to removal by photolysis and volatilization. The volatilization half-life of NDMA from soil surfaces under field conditions has been found to be 1 to 2 hours. In subsurface soil and in water beyond the penetration of sunlight, NDMA would be susceptible to slow microbial decomposition under both aerobic and anaerobic conditions. In aerobic subsurface soil, the half—life of NDMA has been found to be about 50 to 55 days. Degradation has been found to proceed slightly faster under aerobic conditions than under anaerobic conditions. NDMA has been detected in ambient air, water and soil; however, monitoring data are rather scant. Low levels of NDMA (measurable in terms of ppb) are commonly found in the air of car interiors, food, malt beverages (beer, whiskey), toiletry and cosmetic products, rubber baby bottle nipples and pacifiers, tobacco products and tobacco smoke, pesticides used in agriculture, hospitals, and homes, and sewage sludge. The general population is exposed to NDMA from a variety of different sources. Primary sources of exposure include: chewing tobacco, tobacco smoke, foods [beer, liquor, cured meats (particularly bacon), fish, cheeses, and other food items], cosmetics and toiletry articles, interior air of cars, various household commodities such as detergents and home-and-garden pesticides, and formation in the upper gastrointestinal tract during» digestion of secondary amine-containing foods. Infants may also be exposed 66 5. POTENTIAL FOR HUMAN EXPOSURE to NDMA from the use of rubber baby bottle nipples and pacifiers which may contain very low amounts of NDMA, from ingestion of contaminated infant formula, and from breast milk from some nursing mothers. Very low levels of NDMA have been found in breast milk. Occupational settings in which there is potential for exposure to NDMA include, but are not limited to: leather tanneries, rubber and tire industries, rocket fuel industries, dye manufacturers, soap, detergent and surfactant industries, foundries (core- making), fish-processing industries (fish-meal production), pesticide manufacturers, warehouse and sale rooms (especially for rubber products), and research laboratories where NDMA is synthesized/studied. 5.2 RELEASES TO THE ENVIRONMENT 5.2.1 Air NDMA may occasionally be emitted into the atmosphere from sites of manufacture/use of dimethylamine and other sites at which NDMA is inadvertently formed, i.e. tanneries, pesticide manufacturing plants, rubber and tire industries, etc. NDMA may also form in nighttime air as the result of the atmospheric reaction of dimethylamine with N0x (Cohen and Bachman 1978, Fine et al. 1976a, Fine et al. 1976b, Hanst et al. 1977). 5.2.2 Water NDMA may be released in waste streams from facilities at which NDMA was inadvertently formed during manufacturing processes. This would include such facilities as amine manufacturing plants, tanneries, rubber and tire industries, fish processing industries, foundries, rocket fuel industries, dye manufacturers, soap, detergent, and surfactant industries, and pesticide manufacturers (Cohen and Bachman 1978). In addition to industrial sources, NDMA may form in aqueous systems, sewage and soil as the result of either biological, chemical or photochemical processes. Biological formation occurs via the reaction of a secondary or tertiary amine with nitrite. The nitrite can arise in the environment from the microbial transformation of ammonia or nitrate or through manmade production. Chemical formation of nitrosamines occurs optimally under acidic conditions and may occur from the reaction of primary, secondary or tertiary amines with nitrite (Ayanaba and Alexander 1974; Mills and Alexander 1976). Formation of NDMA by photochemical transformation of dimethylamine in the presence of nitrite has been found to occur more readily under alkaline conditions than under acidic or neutral conditions (Ohta et al. 1982). Nitrosamine precursors are widespread throughout the environment, occurring in plants, fish, algae, urine, and feces and are formed in the environment as pesticide degradation products (Ayanaba and Alexander 1974, Greene et al. 1981, Neurath et al. 1977, Windholz 1983). The Contract Laboratory Program statistical data base reports that NDMA has been detected in groundwater samples at one out of 1177 hazardous waste site on the National Priorities List (NPL). This site is Martin Marietta (Denver Aerospace) in Waterton, C0 (VIEW Database 1989). i 67 5. POTENTIAL FOR HUMAN EXPOSURE No data are available regarding contamination of drinking water, irrigation water, sewers, or storm drains in the vicinity of NFL sites. 5.2.3 Soil NDMA may be released into the environment as the result of land application of sewage sludge containing this compound or as the result of land application of certain pesticides contaminated with this compound. NDMA may also form in soils under conditions which favor nitrosation of nitrosamine precursors (Mills and Alexander 1976, Pancholy 1978). There is no data pertaining to the detection of NDMA in soil samples collected at or in the vicinity of NFL sites. 5.3 ENVIRONMENTAL FATE 5.3.1 Transport and Partitioning Organic compounds in the atmosphere having vapor pressures greater than 10' mm Hg are expected to exist almost entirely in the vapor phase (Eisenreich et a1. 1981). The estimated vapor pressure of NDMA [2.7 mm Hg at 20°C (see Table 3-2)] indicates that this compound should not partition from the vapor phase to particulates in the atmosphere. Using linear regression equations based on log Kow data [log Kow = -0.57 (see Table 3-2)], a bioconcentration factor of 0.2 and a soil adsorption coefficient (Koc) of 12 have been estimated for NDMA (Bysshe 1982, Hansch and Leo 1985, Lyman 1982). These values, as well as the complete water solubility of NDMA, indicate that bioaccumulation in aquatic organisms and adsorption to suspended solids and sediments in water would not be important environmental fate processes. The low value of the Henry’s Law Constant for NDMA [2.63x10'7 atm-m3/mol at 20°C (see Table 3-2)] suggests that volatilization would be a relatively insignificant fate process in water (Thomas 1982). NDMA is expected to be highly mobile in soil and it has the potential to leach into groundwater supplies (Dean-Raymond and Alexander 1976, Greene et a1. 1981, Swann et a1. 1983). If NDMA were released to soil surfaces, as might be the case during application of contaminated pesticides, a substantial proportion of the nitrosamine would volatilize. The volatilization half-life from soil surfaces under field conditions is estimated to be on the order of 1-2 hours (Oliver 1979). If NDMA were incorporated into subsurface soil, far less of the nitrosamine would enter the atmosphere by volatilization and the rate of volatilization would be greatly reduced. Under these circumstances volatilization would be of minor importance (Oliver 1979). 68 5. POTENTIAL FOR HUMAN EXPOSURE 5.3.2 Transformation and Degradation 5.3.2.1 Air In the atmosphere, NDMA vapor would rapidly degrade by direct photolysis to form dimethylnitramine. Based on experimental data, the photolytic half—life of NDMA vapor exposed to sunlight has been determined to be about 5 to 30 minutes (Hanst et a1. 1977, Tuazon et a1. 1984). Reaction of NDMA with photochemically-generated hydroxyl radicals or ozone molecules in the atmosphere would be too slow to be environmentally significant (Atkinson and Carter 1984, Tuazon et a1. 1984). 5.3.2.2 Water Limited available data suggest that NDMA would be subject to slow photolysis in natural waters exposed to sunlight (Polo and Chow 1976; Callahan et al. 1979). In unlit waters, it appears that NDMA would be rather persistent, eventually degrading as the result of microbial transformation (Kaplan and Kaplan 1985, Kobayashi and Tchan 1978, Tate and Alexander 1975). There is evidence which suggests that formaldehyde and methylamine may form as biodegradation products of NDMA (Kaplan and Kaplan 1985). Insufficient data are available to predict the rate at which NDMA would degrade in water. NDMA is not expected to chemically react under the conditions found in natural waters (Callahan et a1. 1979, Oliver et a1. 1979). 5.3.2.3 Soil It appears that microbial degradation would be an important removal process for NDMA in subsurface soil. Oliver et a1. (1979) amended Metapeake loam with 10 ppm NDMA at 23°C and observed a half-life of 50 days (Oliver et a1. 1979). Loss of NDMA was attributed to volatilization and biodegradation. Tate and Alexander (1975) amended silt loam with 22.5 ppm NDMA at 30°C and observed a lag of approximately 30 days before slow disappearance from soil commenced; 50% loss occurred after about 55 days incubation and 60% loss occurred after about 70 days incubation. As part of the same study, 40% loss was observed in 2 days in soil amended with 50 ppm NDMA and 44% loss was observed in 5 days in soil amended with 250 ppm NDMA. These initial losses were followed by very little or no loss over the next 3 weeks. Initial, rapid loss of NDMA was attributed to volatilization and slow, gradual loss of NDMA was attributed to biodegradation. Mallik and Tesfai (1981) incubated NDMA at 4, 25 and 37°C and found that at all three _ temperatures, about 20-30% of added NDMA disappeared in the first 20 days of incubation, but little loss was noted thereafter; even after 30 days of incubation, over 50% of the NDMA was retained. The rate of disappearance of NDMA was found to be slightly higher in sandy loam soil than in either clay or silt loam soil. The rate of loss was also found to be slightly higher in aerobic soil at field capacity compared to super saturated (anaerobic) soil. After a 30-day incubation period, 60% of added NDMA remained in soil at 69 5. POTENTIAL FOR HUMAN EXPOSURE field capacity and 70% of added NDMA remained in super saturated soil. Available data on the degradation of NDMA in water and air indicate that photolysis may be an important removal process on soil surfaces. 5.4 LEVELS MONITORED OR ESTIMATED IN THE ENVIRONMENT 5.4.1 Air When it was used as a rocket fuel intermediate, NDMA was identified in ambient air on-site and in the vicinity of factories which were manufacturing rocket fuel (Fine et al. 1977b; Gordon 1978). At a plant in Baltimore, MD, which was manufacturing unsymmetrical dimethylh drazine rocket fuel, the average concentration on-site was 11 600 ng/m , and in neighboring residential communities it was 1,070 ng/mé, with levels ranging between 30 to 100 ng/m3 in the downtown area (Fine et al. 1977b). As a result of these findings, the use of NDMA was discontinued at this plant (Shapley 1976). During December 1975, NDMA was found in air samples collected in Belle, WV near a factory which was manufacturing dimethylamine. The highest level found (980 ng/m3) was collected during a temporary weather inversion (Fine et al. 1976b). NDMA has also been measured in ambient air in urban areas with no known point sources of nitrosamines: Baltimore, MD several miles upwind of the rocket fuel plant (0.02—0.l pg/m3); the Cross Bronx Expressway in New York City (0.8 pg/m3); and Philadelphia, PA (0.025 ppb) (Fine et al. 1976b, Shapley 1976). Occurrence of volatile nitrosamines in air has been associated with tire and rubber products, leather tanneries, and automotive upholstery, and, as a result, measurable levels of the nitrosamines have been found in certain confined areas, e.g. automobile interiors. Concentrations of NDMA in interior air of automobiles have been found to vary widely due to differences in age of the car, design and decor. Levels of NDMA in interior air of new cars were found to range from <0.02 to 0.83 ,ug/m3 (Dropkin 1985, Rounbehler et a1. 1980). 5.4.2 Water Data from the EPA STORET Water Quality data base indicate that NDMA is not a common contaminant of surface waters in the United States (EPA 1988b). During the time when NDMA was being used as a chemical intermediate at a rocket fuel manufacturing plant in Baltimore, MD, concentrations up to 940 ng/L were found in adjacent surface waters. Mud puddles adjacent to the facility contained 0.20-9.0 mg/kg (moist basis) of NDMA (Fine et al. 1977b). Information found in STORET also reveals that NDMA is infrequently found in groundwater samples. STORET gross analysis data input from 1980 to 1988 indicate that NDMA was positively identified in 0.9% of 2308 groundwater samples collected in the United States. The average concentration of positive samples was 12.4 pg/L (EPA 1988b). NDMA also has been detected at a concentration of 10 pg/L in groundwater samples at one of 1177 hazardous waste sites on the National Priorities List (NPL). This site is Martin 7O 5. POTENTIAL FOR HUMAN EXPOSURE Marietta (Denver Aerospace) in Waterton, Co (VIEW 1989, VIAR 1987). NDMA was reportedly found in tap water from Philadelphia, PA at levels of 0.003- 0.006 pg/L (Kimoto et a1. 1981). The authors of this study concluded that NDMA did not form in the resin used to accumulate the nitrosamines, but that it may have formed from the reaction of low concentrations of nitrite, an oxidizing agent (possibly chlorine) and secondary amines present in the water sample. NDMA has been found in deionized laboratory water at levels ranging from 0.03-0.34 pg/L (Fiddler et a1. 1977, Cough et a1. 1977). Anion exchanger resins were identified as the source of NDMA found in the water samples. There have been reports of NDMA occurring infrequently in wastewater samples collected from various locations situated throughout the United States. When present, levels of NDMA are generally in the low pg/L range (maximum reported concentration 2.7 pg/L) (Cohen and Bachman 1978, Ellis et a1. 1982, EPA 1988b, Fine et al. 1977b). 5.4.3 Soil NDMA has been found in soil at 1-8 pg/kg (dry basis) in Belle and Charleston, WV, New Jersey and New York City (Fine et al. 19770). It is speculated that occurrence of NDMA in soil may have arisen from (a) absorption of NDMA in air, (b) absorption of dimethylamine from air and its subsequent N-nitrosation, or (c) from pesticide application. 5.4.4 Other Media N-Nitrosamines are formed in foods by the reaction of secondary and tertiary amines with a nitrosating agent, usually nitrous anhydride, which forms from nitrite in acidic, aqueous solution. NDMA is the most common volatile amine found in food. Food constituents and the physical make—up of the food can affect the extent of nitrosamine formation. Ascorbic acid and sulfur dioxide have been used to inhibit the formation of nitrosamines. NDMA has been found in some processed foods as a result of direct-fire drying; it forms from the nitrosation of amines in drying food by oxides of nitrogen in drying air (Scanlan 1983). Trace levels (usually less than 1 ppb) of NDMA have been found in a variety of foods; however, not all samples of a particular type of food contain detectable levels of NDMA. Table 5-1 lists the levels of NDMA which have been found in food. NDMA may also occur in human breast milk. In a study of 51 samples of breast milk collected from 13 nursing women, NDMA concentrations greater than 0.2 ppb were found in 23.5% of the samples, and the maximum concentration detected was 1.1 ppb (Lakritz and Pensabene 1984). During this study, it was determined that eating a meal containing bacon did not result in increased NDMA levels in milk, although eating a meal containing bacon and a vegetable high in nitrate occasionally resulted in higher levels of NDMA in breast milk. NDMA has been found to occur in a variety of toiletry and cosmetic products, including shampoos, hair conditioners, color toners, shower gels, bath cremes and oils, children's shampoos, children’s bath and health care products, and face tonics, cleansers, and masks. In a study of 145 5. POTENTIAL FOR HUMAN EXPOSURE 71 TABLE 5-1. Detection of N-Nitrosodimethylamine in Fooda Food Item Concentration (pg/kg) Vegetable oils and margarines Apple cider distillates Dried cheeses (parmesan, romano, and American) Soy-containing foods Non-fat dry milk Milk Infant formula Dried legumes Malt vinegar Cereal products Cooked fish Chinese seafood Meat Fried bacon 0.22-1.01 1-10b 0.2-0.3 0.1-0.6 0.17-4.47 0.05-0.60 1 0.2-0.8 0.4 0.3-4.2 0.1-4.2 0.1-131.5 0.1-7.4 1-44 aSources: Canas et a1. 1986, Fazio and Havery 1982, Fiddler et a1. 1981, Goff and Fine 1979, Huang et a1. 1981, Lakritz and Pensabene 1981, Lawrence and Weber 1984, Sen and Seaman 1981b, Sen et a1. 1984, Sen et al. 1985a, Song and Hu 1988, Weston 1984 b #g/L 72 5. POTENTIAL FOR HUMAN EXPOSURE products, 50 samples (34.5%) contained NDMA, with a maximum concentration of 24 pg/kg occurring in a sample of shampoo (Spiegelhalder and Preussman 1984). The U.S. Food and Drug Administration (FDA) established an action level, effective January 1, 1984, of 60 ppb total N-nitrosamines in rubber nipples as measured by a dichloromethane extraction procedure (Thompson et al. 1986). This means that the Consumer Product Safety Commission can take action against any company which introduces baby bottle or pacifier nipples into interstate commerce containing greater than 60 ppb total N-nitrosamines. Compliance testing of infant pacifiers entered into commerce after January 1, 1984 and sold in the U.S. revealed that total N-nitrosamine levels ranged from not detectable to 36.9 ppb, and that NDMA levels ranged from not detectable to 3.55 ppb, with infrequent occurrence of NDMA (Billedeau et al. 1986). This compares well with levels found in pacifiers entered into commerce prior to January 1, 1984, when total N-nitrosamine levels as high as 332 ppb and NDMA levels as high as 6.78 ppb were detected using the same analytical procedure (Billedeau et al. 1986). It should be noted that several companies have discontinued supplying rubber nipples since January 1984, because they could not meet the compliance level. Most malt beverages, regardless of origin, contain NDMA. This includes many domestic and foreign beers and most brands of whiskey (Havery et al. 1981, Hotchkiss et al. 1981, Scanlan et al. 1980, Sen and Seaman 1981C). It is generally accepted that the nitrosamine is formed in malt during the direct-drying phase of its processing (Fazio and Havery 1982). At one time, it was estimated that 64% by weight, of the dietary intake of NDMA of the West German male,population could be attributed to the consumption of beer (Hotchkiss et all 1981, Spiegelhalder et al. 1979). As a result of these findings, the U.S. Food and Drug Administration established an action level of 5 ppb for NDMA in malt beverages sold in the United States (Hotchkiss et a1. 1981). Compliance testing of domestic (United States) and imported beers by the FDA showed that domestic beers (180 samples) contained NDMA levels ranging from not detectable to 9 ppb, with the average level being less than 1 ppb (1% contained greater than 5 ppb), and that imported beers (80 samples) contained levels ranging from not detectable to 13 ppb, with an average level of 1 ppb (5% contained greater than 5 ppb) (Havery et a1. 1981). These results compared favorably with levels found during a market survey carried out prior to establishment of the action level, when 81% of domestic beers contained greater than or equal to 1 ppb and 17% contained greater than 5 ppb (Hotchkiss et al. 1981). Compliance survey data indicate that levels of NDMA in scotch whiskey (44 samples) ranged from not detectable to 2 ppb, with an average of less than 1 ppb (Havery et a1. 1981). NDMA is commonly found in commercially—available tobacco products in the United States. Results of one study showed that chewing tobaccos purchased in the United States contained NDMA at levels ranging from (0.2 to 73 5. POTENTIAL FOR HUMAN EXPOSURE 85.1 ppb (Brunnemann et al. 1985). NDMA also occurs in mainstream and sidestream smoke from cigarettes and other tobacco products, with higher levels occurring in sidestream smoke than in mainstream smoke (Brunnemann et a1. 1983, Chamberlain and Arrendale 1982, McCormick et al. 1973). Sidestream smoke from commercially—available tobacco products purchased in the United States were found to contain NDMA at the following levels: non- filtered cigarette, 680 ng/cigarette; filtered cigarette, 736 ng/cigarette; and small cigar, 1700 ng/cigarette. The ratio of NDMA in sidestream smoke to NDMA in mainstream smoke in the non-filtered cigarette, filtered cigarette and small cigar was found to be 5221, 139:1, and 41:1, respectively (Hoffman et al. 1987). NDMA has been found to occur in various technical and commercial pesticides used in agriculture, hospitals and homes as the result of (a) formation during the manufacturing process, (b) formation during storage, and (c) contamination of amines used in the manufacturing process (Bontoyan et al. 1979). Herbicides in which NDMA has been found include the amine salt formulations of 2,4-D, dicamba, MCPA, MCPP,and 2,3,6-trichlorobenzoic acid. Levels ranging from 0.05 to 640 ppm have been detected in these herbicides (Bontoyan et a1. 1979, Cohen et a1. 1978, Hindle et a1. 1987, Ross et al. 1977). NDMA is a common constituent of municipal sewage sludge (Brewer et a1. 1980, Mumma et al. 1984). NDMA was detected in dried sludges from 14 out of 15 cities geographically located throughout the U.S. at levels ranging from 0.6-45 ppb (Mumma et al. 1984). Occurrence of NDMA in sewage sludge appears to be the result of biological and chemical transformation of alkylamines in the presence of nitrite (Ayanaba and Alexander 1974, Mills and Alexander 1976, Pancholy 1978). 5.5 GENERAL POPULATION AND OCCUPATIONAL EXPOSURE N-Nitrosamine precursors can be found in a large variety of man-made and natural products. Such products include agricultural chemicals, tobacco, detergents, rust inhibitors, rubber additives, solvents, drugs, plastics, leather tanning, textiles, and cosmetics. Considering the widespread occurrence of these products and the common occurrence of nitrogen oxides in industry, there is a fairly high likelihood that N-nitrosamines are found in these products or in industrial setting in which these products are used and/or produced (Fajen 1980). Occupational settings in which there is potential for exposure to NDMA include, but are not limited to: leather tanneries, rubber and tire industries, rocket fuel industries, dye manufacturers, soap, detergent and surfactant industries, foundries (core-making), fish-processing industries (fish-meal production), pesticide manufacturers, and warehouse and sale rooms (especially for rubber products) (Spiegelhalder 1984). When present in workroom air, NDMA levels are typically less than 1 ppb (Fajen et al. 1982). Exposure may result from inhalation or dermal contact. Results of a NIOSH survey carried out between 74 5. POTENTIAL FOR HUMAN EXPOSURE 1981 and 1983 indicate that 747 workers are potentially exposed to NDMA in occupational settings (NIOSH 1988). Laboratory workers handling NDMA could potentially be exposed to the nitrosamine as a result of diffusion through rubber gloves. Walker et a1. (1978) showed that rubber gloves worn in research laboratories do not provide complete protection from dermal exposure to NDMA, because 11.8% of the NDMA contained in a dichloromethane solution was found to diffuse through latex surgical gloves into saline solution, over a period of 20 minutes. Dichloromethane is a common solvent for NDMA. General population exposure to NDMA results from a number of different sources, primarily chewing tobacco, tobacco smoke, foods (beer, cured meats, fish, cheeses, and other food items), cosmetic products, interior air of cars, and various household commodities. Exposure to NDMA may also result from its in vivo formation during digestion in the upper gastrointestinal tract of secondary amine-containing foods or drugs, especially those containing dimethylamine (Groenen et a1. 1980, Magee et a1. 1976, Sakai et al. 1984). Infants may be exposed to NDMA from baby bottle nipples and pacifiers which may contain small amounts of NDMA, from ingestion of contaminated infant formulas, and from breast milk from some nursing mothers. Very low levels of NDMA have been found in breast milk. Based on older estimates of dietary intake in Germany, the Netherlands, and England and on recent data pertaining to occurrence of NDMA in various foods in the U.S., it appears that the average adult dietary intake of NDMA in the U.S. is less than 1 pg per day (Preussmann 1984). Insufficient data are available to predict the average daily intake of NDMA from other sources of exposure. 5.6 POPULATIONS WITH POTENTIALLY HIGH EXPOSURE It appears that those segments of the general population with potentially high exposure to NDMA from exogenous sources would include tobacco smokers and nonsmokers who come in contact with tobacco smoke for extended periods of time, snuff dippers, people who are occupationally exposed, and people who consume large quantities of food known to contain NDMA, beer or whiskey. 5.7 ADEQUACY OF THE DATABASE Section 104 (i) (S) of CERCLA, directs the Administrator of ATSDR (in consultation with the Administrator of EPA and agencies and programs of the Public Health Service) to assess whether adequate information on the health effects of NDMA is available. Where adequate information is not available, ATSDR, in cooperation with the National Toxicology Program (NTP), is required to assure the initiation of a program of research designed to determine these health effects (and techniques for developing methods to determine such health effects). The following discussion highlights the availability, or absence, of exposure and toxicity information applicable to 75 5. POTENTIAL FOR HUMAN EXPOSURE human health assessment. A statement of the relevance of identified data needs is also included. In a separate effort, ATSDR, in collaboration with NTP and EPA, will prioritize data needs across chemicals that have been profiled. 5.7.1 Data Needs Physical and Chemical Properties. Physical and chemical properties are essential for estimating the partitioning of a chemical among environmental media. Many physical and chemical properties are available for NDMA; however, measured values for Koo and Henry’s Law Constant at ambient temperature are not available. Methods for estimating these properties appear to provide relatively close estimates of Koc and Henry's Law Constant. Nevertheless, measured values at environmentally significant temperatures would assist in accurately predicting the fate of this compound in the environment. Environmental Fate. Sufficient data are available to develop a general understanding of the environmental fate of NDMA. Kinetic data regarding photolysis in water and on soil surfaces, biodegradation in water under aerobic and anaerobic conditions, and biodegradation in soil under anaerobic conditions are lacking. Natural water grab sample biodegradation studies and soil metabolism studies carried out in the dark under aerobic and anaerobic conditions would be useful in establishing the persistence of NDMA in the environment. Photolysis studies carried out under simulated environmental conditions in water and soil would be useful in establishing the rate of photolytic degradation, the significance of this process as a removal mechanism, and the products of this reaction in these media. Exposure Levels in Environmental Media. Limited data suggest that NDMA may be found in urban air, but recent comprehensive monitoring data pertaining to the detection of NDMA in ambient air are needed to establish this fact. Occurrence of NDMA in air has been associated with rubber products, leather products, and cigarette smoke and measurable levels of NDMA have been found in car interiors. This information, combined with the fact that NDMA has been found in ambient air at various urban locations, suggests that detectable levels of NDMA exist in the interior air of homes, offices, etc. Studies pertaining to the monitoring of NDMA in indoor air are needed to confirm this supposition. Exposure Levels in Humans. Although numerous studies are available concerning the detection of NDMA in various foods, a market basket study is needed to provide a reliable estimate of the average daily dietary intake of NDMA. Available monitoring data on NDMA need to be evaluated, and estimates of the amount of exposure from each source need to be developed. These data would be useful in establishing the relative importance of each source of intake to overall human exposure and for predicting typical levels of exposure to NDMA. 76 5. POTENTIAL FOR HUMAN EXPOSURE Exposure Registries. Since NDMA occurs most commonly in occupational settings as a result of its inadvertent formation, it would be difficult to develop a reliable estimate of occupational exposure to this compound. Nevertheless, NIOSH has established a registry for occupational exposure to NDMA. It would be difficult to develop a registry for environmental exposure to NDMA since such exposure can occur from a wide variety of sources and level of exposure can vary markedly depending upon an individual's lifestyle. There is no registry available for environmental exposure to this compound. 5.7.2 On-going Studies There is no indication that there are any studies currently in progress which are related to the level of NDMA in environmental media, environmental fate of NDMA, or general population or occupational exposure to NDMA. 77 6. ANALYTICAL METHODS 6.1 BIOLOGICAL MATERIALS Methods used for the quantification of NDMA in biological samples are given in Table 6-1. Two problems encountered in the analysis of NDMA are poor recovery of the compound due to its high volatility and the artifactual formation of this compound during sample storage and treatment (Fine 1982). Since nitroso compounds are formed in acid solution, keeping the solution alkaline during storage and treatment may reduce artifact formation (Kosaka et a1. 1984). Other authors have used ascorbic acid to inhibit in vitro formation of nitrosamines and have used morpholine to measure the extent of in vitro nitrosation during storage and handling (Dunn et al. 1986). The method that has most selectivity for the quantification of this compound is thermal energy analyzer (TEA). A few investigators have oxidized this compound with pentafluoroperoxybenzoic acid to achieve higher sensitivity with electron capture detector (ECD) than with TEA (Kimoto et al. 1984). However, ECD detectors have less selectivity than TEA and will require more sample clean up. The confirmation of NDMA in a sample is usually done by mass spectrometry (MS). Samples containing small amounts of NDMA cannot be detected by MS in the presence of large background impurities (as in samples treated for TEA analysis). Photolysis at 366 nm affords an alternative means for validating the presence of this compound identified by TEA (Cooper et al. 1987). A method for the analysis of total N-nitroso compounds in gastric juice is also available (Pignatelli et al, 1987). 6.2 ENVIRONMENTAL SAMPLES Methods for quantifying NDMA in environmental samples are summarized in Table 6-2. As with the biological samples, in situ artifact formation must be avoided in order to get accurate results from the analysis of environmental samples (Fisher et a1. 1977; Fine et al. 1977a). The three quantification methods that give satisfactory sensitivity for NDMA are alkali flame ionization detector (in the nitrogen mode) (AFID), Hall electrolytic conductivity detector (HECD) in the reductive mode and TEA. The advantages and disadvantages of these detectors have been evaluated (Rhoades et a1. 1980; Usero et al. 1987). Of the three detectors, the TEA detector has the highest sensitivity and selectivity. Because of its higher selectivity, the TEA detector cannot be versatile enough for multipollutant analysis. Mass spectrometric detector can be used not only for confirmation of the presence of NDMA in a sample, but for quantification as well (Eichelberger et a1, 1983; Webb et a1, 1979). When used in combination with a high resolution GC column, this method has the ability to quantify a large number of pollutants in a sample. The use of selected ion monitoring (SIM) may increase the sensitivity by orders of magnitude. The SIM method does not provide the full mass spectra necessary for the identification of unexpected compounds, however (Bellar et a1. 1979). A method for the TABLE 6-1. Analytical Methods for Determining N-Nitrosodimethylamine in Biological Sanples Sanple Matrix Sample Preparation Analytical Method Reference whole blood Blood, liver, kidney, brain Blood Brain, liver, kidney, pancreas Urine Distill alkaline solution, extract dis- tillate in solvent and concentrate. Sample with added sulfamic acid and anti- foaming agent, subjected to simultaneous distillation and extraction, extract con- centrated. Sample mixed with ascorbic acid and mor- pholine, subjected to distillation in alka- line solution, distillate extracted with solvent and concentrated. Alkaline sanple dialyzed with solvent, dialyzates separated and concentrated. Vacuum distillation in mineral oil, extracted with solvent and concentrated. Sample mixed with amnoniun sulfanate, homogenized and distilled under vacuum, extracted with solvent, derivatized with pertrifluoroacetic acid, cleaned by colum chromatography and concentrated. Sample buffered at pH 10 extracted with solvent, solvent concentrated. GC-TEA GC-TEA GC-TEA HRGC-MS GC-TEA GC-TEA and GC-ECD GC'HRMS Detection Accuracy Limit 0.1 ug/L 95% <1 ppb 93-97% at 2.3-4.2 ppb 8 pg or 0.05 93% Its/k9 (for 20 g sanple) 3-4 pg 60-70% 0.1 [Lg/L N6 N6 54.7% 5 ng/L 99-10374 at 10-80 ng/L Lakritz et al. 1980 Pylypiw et al. 1985; Pylypiw 1987 Dunn et al. 1986 Kosaka et al. 1984 cough et al. 1983 Cooper et al. 1987 Garland et al. 1986 N6 = Not given; ct: = gas chromatography; TEA = thermal energy analyses; HRGC = high resolution gas chromatography; MS = [less spectrometry; ECD = electron capture detector; HRMS = high resolution mass spectrometry '9 SGOHLSN TVDILKTVNV 8L TABLE 6-2. Analytical Methods for Determining N-Nitrosodimethylamine in Envirormental Sanples suri mi by colum chromatography and concentrated Sanple Matrix Sample Preparation Analytical Detection Accuracy Reference Method Limit Ambient air Sanple collected in impinger con- GC-MS 10 pg NG Fisher et al. 1977 taining KOH, extracted in solvent and concentrated. Sanple sorbed on Tenax, thermally Cryofocusing 0.5 ppt (for 90-110% Sawicki et al. 1977 desorbed. HRGC-MS 150 L air) Collected in Tenax, thermally desorbed HRGC-MS 0.3 pg NG Webb et al. 1979 and trapped in a cryogenically cooled trap and dissolved in solvent and con- centrated. Collected in aubient or cold KOH trap, GC-TEA 1 ng/m 43.6% Fine et al. 1977a,b extracted in solvent and concentrated. Hater, wastewater Sanple extracted with solvent, colum GC-NPD or 0.15 [lg/L 32% at 0.8 ug/L EPA 1982 chromatographic clean-up, concentration. GC-reductive HECD or GC-TEA (EPA Method 607) Extract with solvent at pH 7, concentrate Cryofocusing 1'10 ug/L 42% at 100 ug/L Eichelberger et al. extract. HRGC-MS (EPA 1983 Method 625.1) Hater Extract with solvent, concentrate extract. GC-TEA 2 ng/L 68% Fine et al. 19778,b Hater, wastewater Extracted with solvent, colum chromato- GC-AFID N6 32% (AFID) Rhoades et al. 1980 graphic clean-q) if required, concentra- GC-TEA 42% (TEA) tion of extract. GC-reduction HECD 43% (HECD) Soil Extracted with water, water extracted GC-TEA NE N6 Fine et al. 1977b with solvent and concentrated. Minced fish and sanple eluted with solvent, cleaned GC-TEA 0.2 ppb 77-97% at 10 ppb Pensabene and Fiddler 1988 '9 S GOHIHN 'IVD I lXIVNV 6L TABLE 6-2 (continued) Sanple Matrix Sauple Preparation Analytical Method Detection Limit Accuracy Reference Meat, vegetable Malt, beer, milk powder, cured meat Malt beverages Beer Fried bacon Vacuun distill gromd sanple, extract distillate with solvent, clean up by colum chromatography, derivatize with peroxytrifluoroacetic acid and colum chromatographic clean up and concentrate. Clean sanple by dry colum/elution or mineral oil distillation method, clean Lp further by colum chromatography, oxidize with pentafluoroperoxybenzoic acid, clean (p by colum chromatography and concentrate. Clean sanple by celite colm'n chromato- graphy, concentrate methylene chloride eluate. Sample treated with sulfanic acid, dis- tillation mder basic condition, extrac— tion with solvent and concentration. Clean sanple by acidic celite colum GC- ECD GC-TEA or GC-ECD GC- TEA GC~ TEA GC- TEA 0.2 ppb (for 250 g sanple) <1ppb "G 0.1 ppb NG <78% "6 90% 78-1121 at 0.08-4 ppb 101% at 10 ppb Telling 1972 Kimoto et al. 1984 Hotchkiss et al. 1981 Sen and Seanan 1981a Pensabene et al. 1982 '9 SGOHLHN TVDILATVNV 08 cc = Gas chromatography; MS = mass spectrometry; HRGC = High resolution gas chromatography; TEA = thermal energy analyzer; NPD = nitrogen- phosphorus detector; MECD = Mall electrolytic conductivity detector; AFID = alkali flame ionization detector; ECD = electron capture detector; N6 = not given 81 6. ANALYTICAL METHODS analysis of apparent total N-nitroso compounds in beer is also available (Massey et a1. 1987). 6.3 ADEQUACY OF THE DATABASE Section 104 (i) (5) of CERCLA, directs the Administrator of ATSDR (in consultation with the Administrator of EPA and agencies and programs of the Public Health Service) to assess whether adequate information on the health effects of NDMA is available. Where adequate information is not available, ATSDR, in cooperation with the National Toxicology Program (NTP), is required to assure the initiation of a program of research designed to determine these health effects (and techniques for developing methods to determine such health effects). The following discussion highlights the availability, or absence, of exposure and toxicity information applicable to human health assessment. A statement of the relevance of identified data needs is also included. In a separate effort, ATSDR, in collaboration with NT? and EPA, will prioritize data needs across chemicals that have been profiled. 6.3.1 Data Needs Methods for Determining Parent Compounds and Metabolites in Biological Materials. NDMA is equally distributed in the cellular elements of the blood and in the plasma and serum (Lakritz et al. 1980). Therefore, it is advantageous to analyze whole blood for the quantification of NDMA. Because NDMA is metabolized almost quantitatively in humans (Spiegelhalder 1984), determination of this compound in human urine needs an extremely sensitive technique (Garland et a1. 1986). The urinary excretion of NDMA has been correlated with the concentration of N02 in air, suggesting that ambient air may play a role in the exposure of people to nitrosoamines (Garland et al. 1986). There is a paucity of data on the analytical methods for the determination of N-nitrosodimethylamine in human urine. No metabolite of NDMA from human exposure to this compound has yet been identified (see Subsection 2.6.3). A metabolite identified in laboratory animal has been discussed in Subsection 2.6.3. The changes in metabolite concentrations with time in human blood, urine, or other appropriate biological medium may be useful in estimating its rate of metabolism in humans. In some instances, a metabolite may be useful in correlating the exposed doses to the human body burden. Such studies on the levels of metabolites in human biological matrices are not available for this compound. Methods for Biomarkers of Exposure Recently, a radioimmunoassay was used to detect elevated levels of the promutagenic lesion O6- methyldeoxyguanosine in DNA cells from individuals with high incidence of cancer who consumed foods with a high nitrosamine content (Wild et a1, 1987). Although no correlation has been established between the DNA-adduct 82 6. ANALYTICAL METHODS and the level of NDMA in consumed foods, the DNA-adduct has the potential to be used as a biomarker for exposure to NDMA. Methods for Determining Parent Compounds and Degradation Products in Environmental Media. The levels of this compound in environmental media can be used to indicate exposure of humans to this compound through the inhalation of air and ingestion of drinking water and foods containing N-nitrosodimethylamine. If a correlation with human tissue or body fluid levels were available, the intake levels from different environmental sources could be used to estimate the body burden of the chemical in humans. Such studies correlating the levels of this compound in any environmental medium with the levels in any human tissue or body fluid are not available. Although the products of biotic and abiotic processes of this compound in the environment are known, no systematic study is available that measured the concentrations of its reaction products in the environment. In instances where the products of an environmental reaction are more toxic than the parent compound, it is important that the level of the reaction products in the environment be known. N-nitrosodimethylamine is not likely to form more toxic products as a result of environmental reactions (see Subsection 5.3.2). The analytical methods for the determination of the levels of environmental reaction products of N-nitrosodimethylamine are available. 6.3.2 On-going Studies No ongoing studies are in progress for the improvement of the analytical method for NDMA in biological samples. Studies are currently conducted by J. Conboy and J. Hotchkiss at Cornell University, Ithaca, NY and by D. Havery at FDA, Washington, DC, for the development of analytical methods for this compound in environmental samples. 83 7. REGULATIONS AND ADVISORIES The International Register of Potentially Toxic Chemicals (IRPTC 1988) lists regulations imposed by 13 countries for NDMA for occupational exposure, packing, storing and transport, disposal, and warns of its probable human carcinogenicity and its high level of toxicity by ingestion or inhalation. NDMA is regulated by effluent guidelines under the Clean Water Act for the following industrial point sources: electroplating, steam electric power generation, asbestos products manufacturing, timber products processing, metal finishing, paving and roofing, paint formulating, ink formulating, and carbon black manufacturing (EPA 1988a). Additional national and state regulations and guidelines pertinent to human exposure to NDMA are summarized in Table 7-1. 84 7. REGULATIONS AND ADVISORIES TABLE 7-1. Regulations and Guidelines Applicable to N-Nitrosodimethylamine Agency Description Value Reference INTERNATIONAL WHO Cancer Classification Group ZAa IARC 1987a NATIONAL Regulations EPA OERR Reportable Quantity 10 lbs EPA 1988a (Proposed 1987) EPA Extremely Hazardous Substance EPA 1987 Emergency Planning and Release 40 CFR 300 and 355 Notification requirenents: Reportable Quantity 1 lb Threshold Planning Quantity 1,000 lbs OSHA Cancer Designation Cancer - suspect agent 29 CFR 1910.1016 (7/1/88) Guidelines a. E q1* (inhalation) 51/rng/kg/day EPA 1988a b. Hater EPA OHRS Ambient Hater Quality Criteria for the EPA 1980 following lifetime increased cancer 45 FR 79318 risk levels: (11/28/80) (with exposure to water, fish _5 and shellfish) 10 6 14.0 ng/L 10:7 1.4 ng/L 10 0.14 ng/L (With exposure to fish and _5 shellfish only) 10-6 160,000 n9/L 10_7 16,000 ng/L 10 1,600 ng/L c Food FDA Action Level for NDMA in barley malt 10 ppb Fed. Reg.1981 46 FR 39218 d Other EPA q1* (oral) 51/ng/kg/day EPA 1988a EPA Cancer Classification Group 328 EPA 19883 ACGIH Cancer Classification Category AZb ACGIH 1989 85 7. REGULATIONS AND ADVISORIES TABLE 7-1 (continued) Agency Desc ri pt i on Value Reference STATE State Acceptable Atrbient Air Concentrations Kansas 0.0018 us/ (annual avg) NATICH 1987 North Carolina 0.0000 us/ (24 hr avg) NATICH 1987 Pennsylvania-Philadelphia 0.0006 ppb 1 yr avg) NATICH 1987 Virginia 3.0000 ml (24 hr avg) NATICH 1987 Kentucky BACTC State of Kentucky 1986 State Acceptable Drinking Hater Concentrations Kansas 0.0014 ug/L FSTRAC 1988 Minnesota 0.014 ug/L FSTRAC 1988 :Probable hunan carcinogen. contribute to overall exposure. Suspected hunan carcinogen. It is noted for NDMA that exposure by the cutaneous route can potentially cBest available control technology. Use of the best available technology to produce the maximum reduction in emissions at a specific emission site is required. 87 8. REFERENCES *Abanobi SE, Farber E, Sarma DSR. 1979. Persistence of DNA damage during development of liver angiosarcoma in rats fed dimethylnitrosamine. Cancer Res 39:1592—1596. *Abe S, Sasaki M. 1982. Induction of sister-chromatid exchanges by indirect mutagens/carcinogens in cultured rat hepatoma and esophageal tumor cells and in Chinese hamster Don cells co-cultivated with rat cells. Mutat Res 93:409- 418. *ACGIH (American Conference of Governmental Industrial Hygienists). 1989. Threshold limit values and biological exposure indices for 1989-1990. Cincinnati, OH: ACGIH, 32, 46-47. *Adair GM, Carver JH. 1983. Induction and expression of mutations at multiple drug-resistance marker loci in chinese hamster ovary cells. Environ Mutagen 5:161-175. *Aleksandrov VA. 1974. [Embryotoxic and transplacental oncogenic action of symmetrical dialkynitrosamines on the progeny of rats.] Bull Exp Biol Med 78:1308-1310. (Russian) *Althoff J, Pour P, Grandjean C, et a1. 1977. Transplacental effects of nitrosamines in Syrian hamsters. 3. Dimethyl- and dipropylnitrosamine. Z Krebsforsch Klin Onkol 90:79-86. *Amacher DE, Paillet SC. 1982. Hamster hepatocyte—mediated activation of procarcinogens to mutagens in the L5178Y/TK mutation assay. Mutat Res 106(2):305-316. *Amacher DE, Paillet SC. 1983. The activation of procarcinogens to mutagens by cultured rat hepatocytes in the L5178Y/TK mutation assay. Mutat Res 113:77-88. *Anderson LM, Giner-Sorolla A, Ebeling D, et al. 1978. Effects of imipramine, nitrite, and dimethylnitrosamine on reproduction in mice. Res Commun Chem Pathol Pharmacol 19:311-327. *Anderson LM, Harrington GW, Pylypiw HM Jr, et a1. 1986. Tissue levels and biological effects of N~nitrosodimethylamine in mice during chronic low or high dose exposure with or without ethanol. Drug Metab Dispos 14:733-739. *Denotes studies cited in text 88 8. REFERENCES Anderson LM, Hagiwara A, Kovatch RM, et a1. 1989. Transplacental initiation of liver, lung, neurogenic, and connective tissue tumors by N-nitroso compounds in mice. Fund Appl Toxicol 12:604—620. *Andrae U, Jahnel P, Greim H. 1979. Induction of DNA repair synthesis in human lymphoblastoid cells by metabolically activated chemicals as short— term test for DNA-damaging compounds [Abstract]. Mutat Res 64:125. *Andrae U, Schwarz LR. 1981. Induction of DNA repair synthesis in isolated rat hepatocytes by 5-diazouracil and other DNA damaging compounds. Cancer Lett 13:187—193. *Arai M, Aoki Y, Nakanishi K, et a1. 1979. Long-term experiment of maximal noncarcinogenic dose of dimethylnitrosamine for carcinogenesis in rats. Gann 70:549-558. *Araki A, Muramatsu M, Matsushima T. 1984. Comparison of mutagenicities of N-nitrosamines on Salmonella typhimurium TAlOO and Escherichia coli WP2 UVRA/PKlOl using rat and hamster liver S9. Gann 75:8-16. *Archer MC, Lee L, Bruce WR. 1982. Analysis and formation of nitrosamines in the human intestine. In: Bartsch H et al., ed. IARC Monograph Series No. 41: N-nitroso compounds: occurrence and biological effects. Lyons, France: WHO, 357-363. *Atkinson R. 1985. Kinetics and mechanisms of the gas—phase reactions of the hydroxyl radical with organic compounds under atmospheric conditions. Chem Rev 85:69-201. *Atkinson R, Carter WPL. 1984. Kinetics and mechanisms of the gas-phase reactions of ozone with organic compounds under atmospheric conditions. Chem Rev 84:437-470. *Ayanaba A, Alexander M. 1974. Transformation of methylamines and formation of a hazardous product, dimethylnitrosamine, in samples of treated sewage and lake water. J Environ Qual 3:83-89. *Bakke JP, Mirsalis JC. 1984. Measurement of unscheduled DNA synthesis (UDS) in fetal and maternal liver following in vivo exposure to genotoxic agents [abstract]. Environ Mutagen 6:446. *Bamborschke S, O’Connor PJ, Margison GP, Maru GB, Kleihues P. 1983. DNA methylation by dimethylnitrosamine in the Mongolian gerbil (meriones unguiculatus): Implications for a non-inducibe hepatic repair system for 06- methylguanine. Cancer Res 43:1306-1311. *Barnes JM, Magee PN. 1954. Some toxic properties of dimethylnitrosamine. Br J Ind Med 11:167-174. 89 8. REFERENCES *Barnes D, Bellin J, DeRosa C, et a1. 1987. Reference dose (RfD): description and use in health risk assessments. Volume I, Appendix A: Integrated risk information system supportive documentation. Washington, DC: US Environmental Protection Agency, Office of Health and Environmental Assessment. EPA/600/8-86/032a. *Bartsch H, Malaveille C, Camus AM et a1. 1980. Validation and comparative studies on 180 chemicals with S. typhimurium strains and V79 Chinese hamster cells in the presence of various metabolizing systems. Mutat Res 76:1-50. *Bauknecht TH, Vogel W, Bayer U, et a1. 1977. Comparative in vivo mutagenicity testing by SCE and micronucleus induction in mouse bone marrow. Human Genet 35:299-307. *Bellar TA, Budde WL, Eichelberger JW. 1979. The identification and measurement of volatile organic compounds in aqueous environmental samples. In: Schuetzle D, ed. ACS Symposium Series No. 94. Monitoring toxic substances. Washington, DC: ACS, 49-62. *Bermudez E, Mirsalis JC, Eales HC. 1982. Detection of DNA damage in primary cultures of rat hepatocytes following in vivo and in vitro exposure to genotoxic agents. Environ Mutagen 42667-679. *Bhattacharyya K. 1965. Fetal and Neonatal Responses to hepatotoxic agents. J Pathol Bacteriol 90:151-161. *Billedeau SM, Thompson HC Jr, Miller BJ, et a1. 1986. Volatile N-nitrosamines in infant pacifiers sold in the United States as determined by gas chromatography/thermal energy analysis. J Assoc Off Anal Chem 69:31- 34. *Blazak W, Stewart B, DiBiasio-Erwin D, et a1. 1985. Induction of sister chromatid exchanges (SCE) by dimethylnitrosamine (DMN) in Chinese hamster cells co-cultured with primary human hepatocytes (PHH) [abstract]. Environ Mutagen. 7:32. *Blount JL, Woodruff RC, Hudson SJ. 1985. Interaction between mobile DNA- element-induced lethal mutations and chemical mutagens in the hybrid dysgenic system of Drosophila melanogaster. Mutat Res 149:33-40. *Bolognesi C, Rossi L, Santi L. 1988. A new method to reveal the genotoxic effects of N-nitrosodimethylamine in pregnant mice. Mutat Res 207257-62. *Bontoyan WR, Law MW, Wright DP. Jr., 1979. Nitrosamines in agricultural and home-use pesticides. J Agr Food Chem 27:631-635. *Bradley MO, Dysart G, Fitzsimmons K, et al. 1982. Measurements by filter elution of DNA single- and double-strand breaks in rat hepatocytes: effects of nitrosamines and gamma-irradiation. Cancer Res 42:2592-2597. 90 8. REFERENCES *Brambilla G, Cavanna M, Pino A, et a1. 1981. Quantitative correlation among DNA damaging potency of six N-nitroso compounds and their potency in inducing tumor growth and bacterial mutations. Carcinogenesis 21425—429. *Brewer WS, Draper AC III, Wey SS. 1980. The detection of dimethylnitrosamine and diethylnitrosamine in municipal sewage sludge applied to agricultural soils. Environ Pollut Ser B Chem Phys 1:37-43. *Brodberg RK, Mitchell MJ, Smith SL, et a1. 1987. Specific reduction of N,N-dimethy1nitrosamine mutagenicity in Drosophila melanogaster by dimethyl sulfoxide. Environ M01 Mutagen 10:425-432. *Brunnemann KD, Masaryk J, Hoffmann D. 1983. Role of tobacco stems in the formation of N—nitrosamines in tobacco and cigarette mainstream and sidestream smoke. J Agric Food Chem 31:1221-1224. *Brunnemann KD, Genoble L, Hoffmann D. 1985. N-Nitrosamine in chewing tobacco: an international comparison. J Agric Food Chem 33:1178-1181. *Bysshe SB. 1982. Bioconcentration factor in aquatic organisms. In: Lyman WJ, Reehl WF, Rosenblatt DH, eds. Handbook of chemical property estimation methods. New York, NY: McGraw Hill Book 00., 5-6 *Callahan MA, Slimak MW, Gabel NW, et a1. 1979. Water-related environmental fate of 129 priority pollutants. Vol. II. Washington, DC: Office of Water Planning and Standards, Office of Water and Waste Management, U.S. EPA. EPA 440/4-79-029b. *Canas BJ, Havery DC, Joe FL Jr, et a1. 1986. Current trends in levels of volatile N-nitrosamines in fried bacon and fried-out bacon fat. J Assoc Off Anal Chem 69:1020-1021. *Carlton WW, Price PS. 1973. Dietary copper and the induction of neoplasms in the rat by acetylaminofluorene and dimethylnitrosamine. Fd Cosmet Toxicol 11:827-840. *Carter RL, Percival WH, Roe FJC. 1969. Exceptional sensitivity of mink to the hepatotoxic effects of dimethylnitrosamine. J Pathol 97:79-88. *Carver JH, Salazar EP, Knize MG, et a1. 1981. Mutation induction at multiple gene loci in Chinese hamster ovary cells: The genetic activity of 15 coded carcinogens and noncarcinogens. In: Evaluation of short-term tests for carcinogens: Report of the International Collaborative program. Prog Mutat Res 1:594-601. *CAS (Chemical Abstract Service). 1988. Registry file. On-line: 8: 08/03/88. 91 8. REFERENCES *Cesarone CF, Bolognesi C, Santi L. 1979. DNA repair synthesis in mice spermatids after treatment with N-methyl-N-nitroso urea and N-N-dimethyl- nitrosamine. Preliminary results. Toxicol 12:183-186. *Cesarone CF, Bolognesi C, Santi L. 1982. Evaluation of damage to DNA after in vivo exposure to different classes of chemicals. Arch Toxicol Suppl 52355-359. *Chamberlain WJ, Arrendale RF. 1982. Rapid method for the analysis of volatile N-nitrosamines in cigarette smoke by glass capillary chromatography. J Chromatog 234:478-481. *Clapp NK, Toya RE Sr. 1970. Effect of cumulative dose and dose rate on dimethylnitrosamine oncogenesis in RF mice. J Nat Cancer Inst 45:495-498. *Clapp NK, Tyndall RL, Otten JA. 1971. Differences in tumor types and organ susceptibility in BALB/c and RF mice following dimethylnitrosamine and diethylnitrosamine. Cancer Res 31;196-198. *Clive D, Johnson KO, Spector JFS, et a1. 1979. Validation and characterization of the L5178Y/TK+/- mouse lymphoma mutagen assay system. Mutat Res 59261-108. *Cohen JB, Bachman JD. 1978. Measurement of environmental nitrosamines. IARC Sci Publ 19:357-372. *Cohen 52, Zweig G, Law M, et a1. 1978. Analytical determination of N-nitroso compounds in pesticides by the United States Environmental . Protection Agency A preliminary study. IARC Sci Publ 19:333-342. *Cole RH, Frederick RE, Healy RP, et al. 1984. Preliminary findings of the priority pollutant monitoring project of the nationwide urban runoff program. J Water Pollut Cont Fed 56:898-908. *Cooper SW, Kimbrough RD. 1980. Acute dimethylnitrosamine poisoning outbreak. J Forensic Sci 25: 874-882. *Cooper SF, Lemoyne C, Gauvreau D. 1987. Identification and quantitation of N-nitrosamines in human postmortem organs. J Anal Toxicol 11:12-18. *Crampton RF. 1980. Carcinogenic dose-related response to nitrosamines. Oncology 37:251-254. *Crygan P, Greim H, Garro AJ, et a1. 1973. Microsomal metabolism of dimethylnitrosamine and the cytochrome P-450 dependency of its activation to a mutagen. Cancer Res 33:2983—2986. ., . '9': 92 8. REFERENCES *Dashman T, Kamm JJ. 1979. Effects of high doses of vitamin E on dimethylnitrosamine hepatotoxicity and drug metabolism in the rat. Biochem Pharmacol 28:1485-1490. *Daugherty JP, Clapp NK. 1976. Studies on nitrosamine metabolism: I. Subcellular distribution of radioactivity in tumor—susceptible tissues of RFM mice following administration of (14C)dimethylnitrosamine. Life Sciences 19:265-271. *De Flora S, Zanacchi P, Camoirano A, et a1. 1984. Genotoxic activity and potency of 135 compounds in the Ames reversion test and in a bacterial DNA- repair test. Mutat Res 133:161-198. *Dean-Raymond D, Alexander M. 1976. Plant uptake and leaching of dimethylnitrosamine. Nature 262:394-396. *Den Engelse L, Hollander CF, Misdorp W. 1974. A sex-dependent difference in the type of tumours induced by dimethyl nitrosamine in the livers of C3HF mice. Eur J Cancer 10:129-135. *Diaz Gomez MI, Swann PF, Magee PN. 1977. The absorption and metabolism in rats of small oral doses of dimethylnitrosamine. Biochem J 164:497-500. *Diaz Gomez MI, Godoy HM, Castro JA. 1981. Further studies on dimethylnitrosamine metabolism, activation and its ability to cause liver injury. Arch Toxicol 47:159-168. *Diaz Gomez MI, Godoy Hm, Villarruel MC, et al. 1983. No response of pigeon liver to dimethylnitrosamine acute effects. Cancer Lett 18:157-162. *Diaz Gomez MI, Tamayo D, Castro JA. 1986. Administration of N—nitrosodimethylamine, N-nitrosopyrrolidine, or N’-nitrosonornicotine to nursing rats: their interactions with liver and kidney nucleic acids from sucklings. J Natl Cancer Inst 76:1133~1136. *Dickins M, Wright K, Phillips M, et al. 1985. Toxicity and mutagenicity of 6 anti-cancer drugs in Chinese hamster V79 cells co—cultured with rat hepatocytes. Mutat Res 157:189-198. *Doolittle DJ, Bermudez E, Working PK, et al. 1984. Measurement of genotoxic activity in multiple tissues following inhalation exposure to dimethylnitrosamine. Mutat Res 1411123-127. *Dreisch FA, Gower M, Munson TO. 1980. Survey of the Huntington and Philadelphia river water supplies for purgeable organic contaminants. Annapolis, MD: U.S. EPA. EPA 903/9-81-003. *Dropkin D. 1985. Sampling of automobile interiors for organic emissions. Report. EPA 600/3-85-008. NITS PB85-172567/GAR, 29 pp. 93 8. REFERENCES *Druckrey H, Preussmann R, Ivankovic S, et a1. 1967. [Organotropic carcinogenic effects of 65 different N-nitroso compounds on BD rats.] Z Krebsforsch 69:103-201. (German) *Dunn SR, Pensabene JW, Simenhoff ML. 1986. Analysis of human blood for volatile N-nitrosoamines by gas chromatography-chemiluminescence detection. J Chromatog 377:35-47. *Eichelberger JW, Kerns EH, Olynyk P, et a1. 1983. Precision and accuracy in the determination of organics in water by fused silica capillary column gas chromatography/mass spectrometry and packed column gas chromatography/mass spectrometry. Anal Chem 55:1471-1479. *Eisenreich SJ, Looney BB, Thornton JD. 1981. Airborne organic contaminants in the Great Lakes ecosystem. Environ Sci Tech 15:30-38. *Ellis DD, Jone CM, Larson RA, et a1. 1982. Organic constituents of mutagenic secondary effluents from wastewater treatment plants. Arch Environ Contam Toxicol. 11:373-382. *EPA. 1977. Computer print-out of non-confidential production data from TSCA inventory. Washington, DC. OPTS, CID. *EPA. 19803. Guidelines and methodology used in the preparation of health effect assessment chapters of the consent decree water criteria documents. US Environmental Protecton Agency. Federal Register 45:79347-79357. *EPA. 1980. Ambient water quality criteria for nitrosamines. Prepared by the Office of Health and Environmental Assessment, Environmental Criteria and Assessment Office, Cincinnati, OH, for the Office of Water Regulations and Standards, Washington, DC. EPA 440/5-80-064. NTIS PB8l-117756, C-48. *EPA. 1982. Test methods. Methods for organic chemical analysis of municipal and industrial wastewater. Method No. 607. Cincinnati, OH: Environmental Monitoring and Support Laboratory, U.S. EPA. *EPA. 1986. Graphical Exposure Modeling System (GEMS). PCCHEM computer program. *EPA. 1987. 40 CFR Parts 300 and 355. Extremely hazardous substances list and threshold planning quantities; emergency planning and release notification requirements. Final rule. Fed Reg 52:13378-13410. *EPA. 1988a. Integrated Risk Information System (IRIS). Risk Estimate for carcinogenicity for N-Nitrosodimethylamine. Online. (Verification data 03/01/88.) Office of Health and Environmental Assessment, Environmental Criteria and Assessment Office, Cincinnati, OH. 94 8. REFERENCES *EPA. 1988b. Storage and Retrieval Water Quality Data Base (STORET) 0n- 1ine: 09/06/88 *Fajen JM. 1980. Industrial hygiene study of workers exposed to nitrosamines. In: Proceedings of the first NCI/EPA/NIOSH collaborative workshop: Progress on joint environmental and occupational cancer studies, May 6-8, Rockville, MD. *Fajen JM, Rounbehler DP, Fine DH. 1982. Summary report on N-nitrosamines in the factory environment. In: Bartsch H et al, ed. IARC Monograph Series No. 41, N-nitroso compounds: occurrence and biological effects. Lyons, France: WHO, 223-230. *Faustman E. 1989. Written communication to ATSDR (September 7, 1989). *Fazio T, Havery DC. 1982. Volatile N-nitrosamines in direct flame-dried processed foods. In: IARC Sci Publ 41:277-286. *Federal Register. 1981. Notice: FDA action level for N-nitrosodimethylamine in barley malt; availability of guide. 46:39218- 39219. *Federal Research in Progress. 1988. On-line: July 1988. Dialog File 165. Dialog Information Services, Inc. *Fiddler W, Pensabene JW, Doerr RC, et a1. 1977. The presence of dimethyl- and diethyl-nitrosamines in deionized water. Fd Cosmet Toxicol 15:441-443. *Fiddler W, Pensabene JW, Kimoto WI. 1981. Investigations of edible oils for volatile nitrosamines. J Food Science 46:603-605. *Fine DH. 1982. Presence of nitrosamines in human beings. In: Magee PN, ed. Banbury Report: Nitrosamines and human cancer, Vol. 12. Cold Spring Harbor, NY, Cold Spring Harbor Laboratory, 271-281. *Fine DH, Rounbehler DP, Belcher NM, et al. 1976a. N-Nitroso compounds: detection in ambient air. Science 19221328-1330. *Fine DH, Rounbehler DP, Pellizzari ED, et al. 1976b. N-Nitrosodimethylamine in air. Bull Environ Cont Tox 15:739-746. *Fine DH, Rounbehler DP, Sawicki E, et al. 1977a. Determination of dimethylnitrosamine in air and water by thermal energy analysis: validation of analytical procedures. Environ Sci Technol 11:577-580. *Fine DH, Rounbehler DP, Rounbehler A, et al. 1977b. Determination of dimethylnitrosamine in air, water, and soil by thermal energy analysis: measurements in Baltimore, MD. Environ Sci Technol 11:581-584. 95 8. REFERENCES *Fine DH, Rounbehler DP, Fan T, Ross R. 1977c. Human exposure to N-nitroso compounds in the environment. In: Hiatt HH, Watson JD, Winsten JA, eds. Origins of human cancer. Book A: Incidence of cancer in humans. Cold Spring Laboratory, 293—307. *Fisher RL, Reiser RW, Lasoski BA. 1977. Determination of sub-microgram per cubic meter levels of N-nitrosodimethylamine in air. Anal Chem 49:1821-1823. *Frank CW, Berry CM. 1981. N-Nitrosamines. In: Clayton GD, Clayton FE, eds. Patty's industrial hygiene and toxicology. 3rd ed. New York, NY: John Wiley & Sons, 3117-3133. *Freund HA. 1937. Clinical manifestations and studies in parenchymatous hepatitis. Ann Int Med 10:1144-1155. *Frezza D, Smith B, Zeiger E. 1983. The intrasanguineous host-mediated assay procedure using Saccharomyces cerevisiae: comparison with two other metabolic activation systems. Mutat Res 108:161-168. *Friedman MA, Sanders V. 1976. Acute toxicity of dimethylnitrosamine in the presence of inhibitors of DMN demethylase. Experientia 32:495-496. *Friedman MA, Staub J. 1976. Inhibition of mouse testicular DNA synthesis by mutagens and carcinogens as a potential simple mammalian assay for mutagenesis. Mutat Res 37:67-76. *FSTRAC (Federal-State Toxicology and Regulatory Alliance Committee). 1988. Summary of State and Federal drinking water standards and guidelines. Chemical Communication Subcommittee, March 10, 17. Fumero S, Mellano D, Peano S, et a1. 1985. Micronucleus induction in rat fetal blood by DMNA directly injected into the fetus [abstract]. Mutat Res 1472295. *Fussgaenger RD, Ditschuneit H. 1980. Lethal exitus of a patient with N-Nitrosodimethylamine poisoning 2.5 years following the first ingestion and signs of intoxication. Oncol 37:273-277. *Garland WA, Kuenzig W, Rubio FU, et a1. 1986. Urinary excretion of nitrosodimethylamine and nitrosoproline in humans: interindividual and intraindividual differences and the effect of administered ascorbic acid and alpha-tocopherol. Cancer Res 46:5392-5400. *Gellert J, Moreno F, Haydn M, et a1. 1980. Decreased hepatotoxicity of dimethylnitrosamine (DMN) following chronic alcohol consumption. Adv Exp med Biol 132:237-243. *Geyer H, Scheunert I, Korte F. 1986. Bioconcentration potential of organic environmental chemicals in humans. Regul Toxicol Pharmacol 62313-347. 96 8. REFERENCES *Goff EU, Fine DH. 1979. Analysis of volatile N-nitrosamines in alcoholic beverages. Fd Cosmet Toxicol 17:569-573. *Gordon RJ. 1978. Survey for airborne nitrosamines [Abstract]. Gov Rep Announce Index 79:65. *Gough TA, Webb KS, McPhail MF. 1977. Volatile nitrosamines from ion- exchange resins. Fd Cosmet Toxicol 15:437-440. *Gough TA, Webb KS, Swann PF. 1983. An examination of human blood for the presence of volatile nitrosamines. Fd Chem Toxicol 21:151-156. *Greene S, Alexander M, Leggett D. 1981. Formation of N—nitrosodimethylamine during treatment of municipal waste water by simulated land application. J Environ Qual 10:416-421. *Griciute L, Castegnaro M, Bereziat JC. 1981. Influence of ethyl alcohol on carcinogenesis with N-nitrosodimethylamine. Cancer Lett 13:345-352. *Groenen PJ, de Cock-Bethbeder MW, Bouman J. et a1. 1980. Formation of N-nitrosamines and N-nitrosamino acids from food products and nitrite under simulated gastric conditions. Proc of the VIth Intern Symp on N-Nitroso Compounds International Agency for Research on Cancer (IARC) Lyon. *Hadjiolov D, Mundt D. 1974. Effect of aminoacetonitrile on the metabolism of dimethylnitrosamine and methylation of RNA during liver carcinogenesis. J Natl Cancer Inst 52:753-756. *Hamilton A, Hardy HL. 1974. Industrial Toxicology. Third Edition Publishing Science Group, Inc. Acton, MA. p. 311. *Hansch C, Leo AJ. 1985. Medchem project issue no. 26. Claremont, CA, Pomona College. *Hanst PL, Spence JW, Miller M. 1977. Atmospheric chemistry of N-nitrosodimethylamine. Environ Sci Technol 11:403-405. *Hard GC, Butler WH. 1970. Toxicity of dimethylnitrosamine for the rat testis. J Pathol 102:201-207. *Hashimoto S, Yokokura T, Kawai Y, et a1. 1976. Dimethylnitrosamine formation in the gastro-intestinal tract of rats. Fd Cosmet Toxicol 14:553- 556. *Havery DC, Hotchkiss JH, Fazio T. 1981. Nitrosamines in malt and malt beverages. J Food Sci 46:501-505. 97 8. REFERENCES *Havery DC, Perpetti GA, Canas BJ, et a1. 1985. Reduction in levels of volatile N-nitrosamines in rubber nipples for babies' bottles. Fd Chem Toxicol 23:991-993. . *Hawley CG. 1981. The condensed chemical dictionary. 10th ed. New York, NY. Van Nostrand Reinhold Company, Inc., 737. *Heading CE, Phillips JC, Lake BC et a1. 1974. Some aspects of the metabolism of dimethylnitrosamine in the rat. Biochem Soc Trans 2:607-610. *Heath DF. 1962. The decomposition and toxicity of dialkyinitrosamines in rats. Biochem J 85:72-91. *Hemminki K. 1982. Dimethylnitrosamine adducts excreted in rat urine. Chem- Biol Interact 39:139—148. *Herron DC, Shank RC. 1980. Methylated purines in human liver DNA after probable dimethylnitrosamine poisoning. Cancer Res 40:3116-3117. *Hindle RW, Armstrong JF, Peake AA. 1937. Determination of N-nitrosodimethylamine levels in some Canadian 2,4-D amine formulations. J Assoc Off Anal chem 70:49-51. *Hoffman D,Adams JD, Brunnemann KD. 1987. A critical look at N-nitrosamines in environmental tobacco smoke. Toxicol Lett 35:1-8. *Holsapple MP, Munson AE, McNerney P, et a1. 1983. Immunosuppression by subchronic exposure to dimethylnitrosamine in female B6C3F1 mice. In: 67th annual meeting of the Federation of American Societies for Experimental Biology, Chicago, IL, April 10-15. Fed Proc 421abstract 469. *Holsapple MP, Tucker AN, McNerney PJ, et a1. 1984. Effects of N-nitrosodimethylamine on humoral immunity. J Pharmacol Exp Ther 229:493- 500. *Holsapple MP, Bick PH, Duke SS. 1985. Effects of N-nitrosodimethylamine on cell-mediated immunity. J Leukocyte Biol 37:367-381. *Hotchkiss JH, Havery DC, Fazio T. 1981. Rapid method for estimation of N-nitrosodimethylamine in malt beverages. J Assoc Off Anal Chem 64:929- 932. *HSDB. 1988. Hazardous Substance Data Base. Record no. 1667. On-line 08/19/88. *Hsie AW, Machanoff R, Couch DB, et a1. 1978. Mutagenicity of dimethylnitrosamine and ethyl methanesulfonate as determined by the host- mediated CHO/HGPRT assay. Mutat Res 51:77—84. 98 8. REFERENCES *Huang DP, Ho, JHC, Webb KS, et al. 1981. Volatile nitrosamines in salt- preserved fish before and after cooking. Fd Cosmet Toxicol. 19:167-172. *IARC (International Agency for Research on Cancer). 1978. IARC monographs on the evaluation of the carcinogenic risk of chemicals to humans. Some N-nitroso compounds Vol. 17. p. 125-175. *IARC (International Agency for Research on Cancer). 1982. Laboratory decontamination and destruction of carcinogens in laboratory wastes: Some N-nitrosoamines. IARC Scientific Publication No. 43. IARC (International Agency for Research on Cancer). 1984. N-nitroso compounds: Occurrence, biological effects and relevance to human cancer. IARC Scientific Publication No. 57. *IARC (International Agency for Research on Cancer). 1987a. IARC monographs on the evaluation of carcinogenic risks to humans. Update of IARC Monographs Vols 1-42, Suppl 7, 31-32, 67-68. IARC (International Agency for Research on Cancer). 1987b. The relevance of N-nitroso compounds to human cancer. Exposures and mechanisms. IARC Scientific Publication No. 84. *Ikeuchi T, Sasaki M. 1981. Differential inducibility of chromosome aberrations and sister-chromatid exchanges by indirect mutagens in various mammalian cell lines. Mutat Res 90:149-161. *Imbriani M, Ghittori S, Pezzagno G, et a1. 1985. Urine/air partition coefficients for some industrially important substances. G Ital Med Lav 7;133-40. *Inoue K, Shibata T, Abe T. 1983. Induction of sister-chromatid exchanges in human lymphocytes by indirect carcinogens with and without metabolic activation. Mutat Res 117:301-309. *Inui N, Nishi Y, Taketomi M, et al. 1979. Transplacental action of sodium nitrite on embryonic cells of Syrian golden hamster. Mutat Res 66:149-158. *Ireton HJC, McGiven AR, Davies DJ. 1972. Renal mesenchymal tumour induced in rats by dimethylnitrosamine: light- and electron-microscope studies. Path 1081181. *IRPTC (International Register of Potentially Toxic Chemicals). 1988. United Nations Environment Programme. Geneva, Switzerland: IRPTC/UNEP. *Ishidate M. Jr., Yoshikawa K. 1980. Chromosome aberration tests with Chinese hamster cells in vitro with and without metabolic activation: A comparative study on mutagens and carcinogens. In: Further studies in the assessment of toxic actions. Arch Toxicol Suppl 4:41-44. 99 8. REFERENCES *Ishinishi N, Tanaka A, Hisanaga A, et a1. 1988. Comparative-study on the carcinogenicity of N-nitrosodiethylamine, N-nitrosodimethylamine, N-nitrosomorpholine, N-nitrosopyrrolidine and N-nitrosodi-n-propylamine to the lung of Syrian golden hamsters following intermittent instillations to the trachea. Carcinogenesis 92947-950. *Ishiwata H, Mizushiro H, Tanimura A, et a1. 1977. Metabolic fate of the precursors of N-nitroso compounds. I. Castro-intestinal absorption of N-nitrosodimethylamine and its precursors in guinea-pigs. J Fd Hyg Soc 18:524-528. *Ito N, Fukushima S, Tsuda H, et a1. 1982. Induction of preneoplastic and neoplastic lesions in rats treated N-nitroso compounds. IARC Sci Publ 41:597-601. *Iversen OH. 1980. Tumorigenicity of N-nitroso—diethyl, -dimethy1 and - diphenyl-amines in skin painting experiments. A study utilizing the tetrazolium test and skin applications on hairless mice. Eur J Cancer 16:695-698. *Jacobson KH, Wheelwright HJ Jr, Clem JH, et a1. 1955. Studies on the toxicology of N-nitrosodimethylamine vapor. AMA Arch Indust. Health 12: 617-622. *Jagannath DR, Vultaggio DM, Brusick DJ. 1981. Genetic activity of 42 coded compounds in the mitotic gene conversion assay using Saccharomyces cerevisiae strain D4. In: Evaluation of short-term tests for carcinogens: Report of the International collaborative Program. Prog Mutat Res 1:456-467. *Jenkins SA, Grandison A, Baxter JN, et a1. 1985. A dimethylnitros- amine- induced model of cirrhosis and portal hypertension in the rat. J Hepatol 1:489-499. *Johansson EB, Tjalve H. 1978. The distribution of (l4C)dimethylnitrosamine in mice. Autoradiographic studies in mice with inhibited and noninhibited dimethylnitrosamine metabolism and a comparison with the distribution of (lAC)formaldehyde. Toxicol Appl Pharmacol 45:565-575. *Johnson KW, Munson AE, Holsapple MP. 1987a. Primary cellular target responsible for dimethylnitrosamine-induced immunosuppression in the mouse. Immunopharmacology 13:47-60. *Johnson KW, Munson AE, Kim D-H, et al. 1987b. Role of reactive metabolites in suppression of humoral immunity by N-nitrosodimethylamine. J. Pharmacol Exp Ther 240:847-855. 100 8. REFERENCES *Kaminski NE, Jordan SD, Page D., et a1. 1989. Suppression of humoral immune responses by dialkylnitrosamines: Structure-activity relationships. Fund Appl Toxicol 12:321-332. *Kaplan DL, Kaplan AM. 1985. Biodegradation of N-nitrosodimethylamine in aqueous and soil systems. Appl Environ Microbiol 50:1077-1086. *Katoh Y, Tanaka M, Takayama S. 1982. Higher efficiency of hamster hepatocytes than rat hepatocytes for detecting dimethylnitrosamine and diethylnitrosamine in hepatocyte-mediated Chinese hamster V79 cell mutagenesis assay. Mutat Res 105:265—269. *Keefer LK. 1989. Written communication (May 1) to Stephen Bosch, Syracuse Research Corporation, Syracuse, NY, regarding on-going metabolism studies. Department of Health and Human Services, Public Health Service, National Institutes of Health, National Cancer Institute, Frederick Cancer Research Facility, Frederick, MD. *Keefer LK, Lijinsky W, Garcia H. 1973. Deuterium isotope effect on the carcinogenicity of dimethylnitrosamine in rat liver. J Natl Cancer Inst 51:299-302. *Keefer LK, Anjo T, Heur YH, et a1. 1987. Potential for metabolic deactivation of carcinogenic N-nitrosodimethylamine in vivo. IARC Sci Publ 84:113-116. *Khanna SD, Puri D. 1966. The hepatotoxic effects of dimethylnitrosamine in the rat. J Path Bact 91:605-608. *Khudoley V, Malaveille C, Bartsch H. 1981. Mutagenicity studies in Salmonella typhimurium on some carcinogenic N-nitrosoamines in vitro and in the host-mediated assay in rats. Cancer Res 41:3205-3210. *Kimbrough RD. 1982. Pathological changes in human beings acutely poisoned by dimethylnitrosamine. In: Magee PN, ed. Banbury Report, Vol. 12. Nitrosamines and human cancer; meeting. Cold Spring Harbor, NY: Cold Spring Harbor Laboratory, 25-36. *Kimoto WI, Dooley CJ, Carre J, et a1. 1981. Nitrosamines in tap water after concentration by a carbonaceous adsorbent. Water Res 15:1099-1106. *Kimoto WI, Silbert LS, Fiddler W. 1984. Confirmation of N-nitrosodimethylamine and N-nitrosopyrrolidine in foods by conversion to their nitramines with pentafluoroperoxybenzoic acid. J Assoc Off Anal Chem 67:751-756. ‘ *Klein RG. 1982. Calculations and measurements on the volatility of N-nitrosoamines and their aqueous solutions. Toxicology 23:135-147. 101 8. REFERENCES *Klein RG, Schmezer P. 1984. Quantitative measurement of the exhalation rate of volatile N-nitrosamines in inhalation experiments with anesthetized Sprague-Dawley rats. IARC Sci Publ 57:513-517. *Kobayashi M, Tchan YT. 1978. Formation of dimethylnitrosamine in polluted environment and the role of photosynthetic bacteria. Water Res 12:199-201. *Koppang N, Rimeslatten H. 1976. Toxic and carcinogenic effects of nitrosodimethylamine in mink. IARC Sci Publ 14:443—452. *Kornbrust D, Dietz D. 1985. Aroclor 1254 pretreatment effects on DNA repair in rat hepatocytes elicited by in vivo and in vitro exposure to various chemicals. Environ Mutagen 71857-870. *Korsrud GO, Grice HG, Goodman TK, et a1. 1973. Sensitivity of several serum enzymes for the detection of thioacetamide, dimethylnitrosamine-and diethanolamine-induced liver damage in rats. Tox Appl Pharm 26:299-313. *Kosaka H, Uozumi M. Nakajima T. 1984. Measurement of nitrosodimethylamine by capillary gas chromatography-mass spectrometry with the 15N—1abe11ed compound as an internal standard. Int Arch Occ Environ Health 54:233-239. *Kroeger-Koepke MB, Michejda CJ. 1979. Evidence for several demethylase enzymes in the oxidation of dimethylnitrosamine and phenylmethylnitrosamine by rat liver fractions. Cancer Res 39:1587-1591. *Kuroki T, Drevon C, Montesano R. 1977. Microsome-mediated mutagenesis in V79 Chinese hamster cells by various nitrosamines. Cancer Res 37;1044-1050. *Lakritz L, Simenhoff ML, Dunn SR, et a1. 1980. N-Nitrosodimethylamine in human blood. Fd Cosmet Toxicol 18:77—79. *Lakritz L, Pensabene JW. 1981. Survey of fluid and nonfat dry milks for N-nitrosamines. J Dairy Sci 64:371-374. *Lakritz L, Pensabene JW. 1984. Survey of human milk for volatile N-nitrosamines and the influence of diet on their formation. Fd Chem Toxicol 22:721—724. *Landon EJ, Naukam RJ, Rama Sastry BV. 1986. Effects of calcium channel blocking agents on calcium and centrilobular necrosis in the liver of rats treated with hepatotoxic agents. Biochem Pharmacol 35:697-705. *Langenbach R. 1986. Mutagenic activity and structure—activity relationships of short-chain dialkyl N—nitrosamines in a hamster hepatocyte V79 cell-mediated system. Mutat Res 163:303—311. 102 8. REFERENCES *Langenbach R, Leavitt S, Hix C, et al. 1986. Rat and hamster hepatocyte- mediated induction of SCEs and mutation in V79 cells and mutation of Salmonella by aminofluorene and dimethylnitrosamine. Mutat Res 161:29-37. *Lawrence JF, Weber DF. 1984. Determination of polycyclic aromatic hydrocarbons in Canadian samples of processed vegetable and dairy products by liquid chromatography with fluorescence detection. J Agric Food Chem 32:794-797. *Lee WR, Abrahamson S, Valencia R. et a1. 1983. The sex-linked recessive lethal test for mutagenesis in Drosophila melanogaster. A report of the U.S. Environmental Protection Agency Genetox Program. Mut Res 123:183-279. *Lele PS, Dunn SR, Simenhoff ML, et a1. 1983. Evidence for generation of the precarcinogen nitrosodimethylamine in the small intestine in chronic renal failure. Kidney Int 24 suppl 16:5229-8233. *Lele P, Dunn SR. Simenhoff ML. 1985. Demonstration of the carcinogen dimetylnitrosamine (DMNA) generation in gut by ethanol inhibition of its first-pass metabolism by liver in chronic renal failure [abstract]. Kidney Int 27:245. *Lele PS, Dunn SR, Simenhoff ML. 1987. Nutritional and metabolic modulation of the carcinogen nitrosodimethylamine in chronic renal failure. Kidney Int 32, suppl 22:S-159-S-l61. *Lijinsky W, Reuber MD. 1981. Comparative carcinogenesis by some aliphatic nitrosamines in Fischer rats. Cancer Lett 14:297-302. *Lijinsky W. 1987; Structure-activity relations in carcinogens by N-nitroso compounds. Cancer Metastasis Rev 6:301-356. *Lijinsky W, Kovatch RM, Riggs CW. 1987. Carcinogenesis by nitrosodialkylamines and azoxyalkanes given by gavage to rats and hamsters. Cancer Res. 47:3968—3972. *Lijinsky W, Reuber MD. 1984. Carcinogenesis in rats by nitrosodimethylamine and other nitrosomethylalkylamines at low doses. Cancer Lett 22:83-88. Lijinsky W. 1986. The significance of N-nitroso compounds on environmental carcinogens. J Environ Sci Health C4(l):1-45. *Lorr NA, Miller KW, Chung HR, et al. 1984. Potentiation of the hepatotoxicity of N-nitrosodimethylamine by fasting, diabetes, acetone, and isopropanol. Toxicol Appl Pharmacol 73:423-431. 103 8. REFERENCES *Lotlikar PD, Baldy WJ Jr, Dwyer EN. 1975. Dimethylnitrosamine demethylation by reconstituted liver microsomal cytochrome P-450 enzyme system. Biochem J 152:705-708. *Lotlikar PD, Hong YS, Baldy WS Jr. 1978. Effect of dimethylnitrosamine concentration on its demethylation by liver microsomes from control and 3- methylcholanthrene pretreated rats, hamster and guinea pigs. Cancer Lett 4:355-361. Lu SH, Yang WX, Guo LP, et a1. 1987. Determination of N-nitrosamines in gastric juice and urine and a comparison of endogenous formation of N-nitrosoproline and its inhibition in subjects from highoand low-risk areas for oesophageal cancer. IARC Sci Publ 84:538-543. *Luster MI, Blank JA, Dean JH. 1987. Molecular and cellular basis of chemically induced immunotoxicity. Ann Rev Pharmacol Toxicol 27:23-49. *Luster MI, Munson AE, Thomas PT, et a1. 1988. Method Evaluation. Development of a testing battery to assess chemical-induced immunotoxicity National Toxicology Program’s guidelines for immunotoxicity evaluation in mice. Fund Appl Toxicol 10:2-19. *Lyman WJ. 1985. Estimation of physical properties. In: Neely WB, Blau GE, eds. Environmental exposure from chemicals, Vol. 1, Boca Raton, FL: CRC Press Inc., 30-31. *Lyman WJ. 1982. Adsorption coefficient for soils and sediments. In: Lyman WJ, Reehl WF, Rosenblatt DH, eds. Handbook of chemical property estimation methods. New York, NY: McGraw-Hill Book Co., 4-9 *Madle E, Kasper P, Madle S, et a1. 1987. Hepatocyte-mediated SCE induction by indirect mutagens: Importance of hepatocyte density and cell—to-cell contact. Mutat Res 188:153-160. ' *Maduagwu EN, Bassir O. 1980. A comparative assessment of toxic effects of dimethylnitrosamine in six different species. Toxicol Appl Pharmacol 53:211-219. *Magee PN. 1956. Toxic liver injury: The metabolism of dimethylnitrosamine. Biochem J 64:676-682. *Magee PN, Barnes JM. 1956. The production of malignant primary hepatic tumours in the rat by feeding dimethylnitrosamine Br J Cancer 10:114-122. *Magee PN, Montesan R, Preussman R. 1976. N-nitroso compounds and related carcinogens. ACS Monograph 1732491-625. 104 8. REFERENCES *Maling HM, Stripp B, Sipes IG, et a1. 1975. Enhanced hepatotoxicity of carbon tetrachloride, thioacetamide, and dimethylnitrosamine by pretreatment of rats with ethanol and some comparisons with potentiation by isopropanol. Toxicol Appl Pharmacol 33:291-308. *Mallik MAB, Tesfai K. 1981. Transformation of nitrosamines in soil and in vitro by soil microorganisms. Bull Environ Contam Toxicol 27:115-121. *Martelli A, Robbiano L, Giuliano L, et al. 1985. DNA fragmentation by N-nitrosodimethylamine and methyl methanesulfonate in human hepatocyte primary cultures. Mutat Res 1442209-211. *Martino PE, Diaz Gomez MI, Tamayo D, et a1. 1988. Studies on the mechanism of the acute and carcinogenic effects of N-nitrosodimethylamine on mink liver. J Toxicol Environ Health 23:183-192. *Massey RC, Key PE, McWeeny DJ.,Jr. et a1. 1987. An investigation of apparent total N-nitroso compounds in beer. In: Relevance of N-nitroso compounds to Human Cancer. Exposures and Mechanisms. IARC Sci Publ 84:219- 221. *Matsuoka A, Hayashi M, Ishidate M., Jr. 1979. Chromosomal aberration tests on 29 chemicals combined with S9 mix in vitro. Mutat Res 66:277-290. *Matsuoka A, Sofuni T, Ishidate M. 1986. Effect of surfactants on the induction of chromosomal aberrations in Chinese hamster cells in culture [Abstract]. Mutat Res 1642273-274. *McCormick A, Nicholson MJ, Baylis MA, et al. 1973. Nitrosamines in cigarette smoke condensate. Nature 244:237-238. *McGiven AR, Ireton HJC. 1972. Renal epithelial dysplasia and neoplasia in rats given dimethylnitrosamine. J Pathol 108:187-190. *McQueen CA, Kreiser DM, Williams GM. 1983. The hepatocyte primary culture/DNA repair assay using mouse or hamster hepatocytes. Environ Mutagen 521-8. *Mehta R, Silinskas KC, Zucker PF, et al. 1987. Micronucleus formation induced in rat liver and esophagus by nitrosamines. Cancer Lett 35:313- 320. *Mills AL, Alexander M. 1976. Factors affecting dimethylnitrosamine formation in samples of soil and water. J Environ Qual 5:437-440. 105 8. REFERENCES *Mirvish SS, Issenberg P, Sornson HC. 1976. Air-water and ether-water distribution of N-nitroso Compound: Implications for laboratory safety, analytic methodology, and carcinogenicity for the rat esophagus, nose, and liver. J Natl Cancer Inst 56:1125-1129. *Moiseev GE, Benemanskii V. 1975. [Concerning the carcinogenic activity of low concentrations of nitrosodimethylamine during inhalation.] Vopr Onkol (Leningr) 21:107-109. (Russian) *Montesano R, Bresil H, Pegg AE. 1982. Metabolism of dimethylnitrosamine and repair of 0-6-methylguanine in DNA by human liver. In: Magee PN, ed. Banbury report, Vol. 12, Nitrosamines and human cancer. Cold Spring Harbor, NY: Cold Spring Harbor Laboratory, 141-152. *Mufti SI, Salvagnini M, Lieber CS, et a1. 1988. Chronic ethanol consumption inhibits repair of dimethylnitrosamine induced DNA alkylation. Biochem Biophys Res Com 152:423-441. *Mumma R0, Raupach DC, Waldman JP, et a1. 1984. National survey of elements and other constituents in municipal sewage sludges. Arch Environ Contam Toxicol 13:75-83. *Myers MJ, Pullen JK, Schook LB. 1986. Alteration of macrophage differentiation into accessory and effector cells from exposure to dimethylnitrosamine (DMN) in vivo. Immunopharmacology 12:105-115. *Myers MJ, Dickens CS, Schook LB. 1987. Alteration of macrophage anti- tumor activity and transferrin receptor expression by exposure to dimethylnitrosamine in vivo. Immunopharmacology 13:195-205. *Napalkov NP, Alexandrov VA. 1968. On the effects of blastomogenic substances during embryogenesis. Z Krebsforsch 71:32-50. *NATICH (National Air Toxics Information Clearinghouse). 1987. NATICH data base report on state, local and EPA air toxics activities, 101. *Neal SB, Probst GS. 1983. Chemically-induced sister chromatid exchange in vivo in bone marrow of Chinese hamsters. An evaluation of 24 compounds. Mutat Res 113233-43. *Neurath GB, Duenger M, Pein FG, et a1. 1977. Primary and secondary amines in the human environment. Fd Cosmet Toxicol 15:275-282. *Ng WL, Fong LYY, Newberne PM. 1984. Forestomach squamous papillomas in the rat: effect of dietary zinc deficiency on induction. Cancer Lett 22:329-332. *NIOSH. 1988. National Occupational Exposure Survey (NOES) as of May 10, 1988. Washington, DC: National Institute for Occupational Safety and Health. 106 8. REFERENCES *Nishie K. 1983. Comparison of the effects of N-nitrosodimethylamine on pregnant and nonpregnant Holtzman rats. Fd Chem Toxicol 21:453-462. *Nishie K, Norred WP, Wasserman A, et a1. 1972. Phototoxicity and differential hepatotoxicity as biological indicators of nitrosamine activity. Toxicol Appl Pharmacol 23:680-691. *O'Connor PJ, Chu Y-H, Cooper DP, Maru GB, Smith RA Species difference in the inducibility of hepatic O rodents. Biochimie 64:769—773. Margison GP. 1982. -alky1guanine repair in 6 *Odagiri Y, Adachi S, Katayama H, et a1. 1986. Detection of the cytogenetic effect of inhaled aerosols by the micronucleus test. Mutat Res 170:79-83. *O’Neill JP, Machanoff R, San Sebastian JR, et a1. 1982. Cytotoxicity and mutagenicity of dimethylnitrosamine in mammalian cells (CHO/HGPRT system): Enhancement by calcium phosphate. Environ Mutagen 4:7-18. *OHM-TADS. (Oil and Hazardous Materials Technical Assistance Data System). 1988. Computer data base. On-line: July 31, 1988. *Ohta T, Suzuki J, Iwano Y, et a1. 1982. Photochemical nitrosation of dimethylamine in aqueous solution containing nitrite. Chemosphere 11:797- 801. *Okinaka RT, Barnhart BJ, Chen DJ. 1981. Comparison between sister- chromatid exchange and mutagenicity following exogenous metabolic activation of promutagens. Mutat Res 91:57-61. *Oliver JE. 1979. Volatilization of some herbicide—related nitrosamines from soils. J Environ Qual 82596-601. *Oliver JE, Kearney PC, Kontson A. 1979. Degradation of herbicide-related nitrosamines in aerobic soils. J. Agric Food Chem 27:887—891. *Omori Y, Takahashi A, Ohno Y, et a1. 1979. Drug-nitrite interactions: Species and sex differences in the formation of dimethylnitrosamine and its effects on the hepatic enzyme activities. Arch Toxicol Suppl 2:349-354. *Otsuka H, Kuwahara A. 1971. Hemangiomatous lesions of mice treated with nitrosodimethylamine. Gann 62:147-156. *Page GW. 1981. Comparison of groundwater and surface water for patterns and levels of contamination by toxic substances. Environ Sci Technol 15:1475- 1481. *Pancholy SK. 1978. Formation of carcinogenic nitrosamines in soils. Soil Biol Biochem 10:27-32. 107 8. REFERENCES *Pedal I, Besserer K, Goerttler K, et a1. 1982. Fatal nitrosamine poisoning. Arch Toxicol 50:101-112. *Pegg AE. 1980. Metabolism of N-Nitrosodimethylamine. IARC Sci Publ 27:3-22. *Pegg AE, Hui G. 1978. Removal of methylated purines from rat liver DNA after administration of dimethylnitrosamine. Cancer Res 38:2011—2017. *Pegg AE, Perry W. 1981. Alkylation of nucleic acids and metabolism of small doses of dimethylnitrosamine in the rat. Cancer Res 41:3128—3132. *Pegg AE, Perry W, Bennett RA. 1981. Effect of partial hepatectomy on removal of 06-methy1guanine from alkylated DNA by rat liver extracts. Biochem J 197:195-201. *Pensabene JW, Miller AJ, Greenfield EL, et a1. 1982. Rapid dry column method for determination of N-nitrosopyrrolidine in fried bacon. J Assoc Off Anal Chem 65:151-156. ' *Pensabene JW, Fiddler W. 1988. Determination of volatile N—nitro— amine in frankfurters containing minced fish and surimi. J Assoc Off Anal Chem 71:839-843. *Peto R, Gray R, Brantom P, et a1. 1984. Nitrosamine carcinogenesis in 5120 rodents: Chronic administration of sixteen different concentrations of NDEA, NDMA, NPYR and NPIP in the water of 4440 inbred rats with parallel studies on NDEA alone of the effect of age of starting (3, 6 or 20 weeks) and of species (rats, mice or hamsters). IARC Sci Publ 57:627-666. *Petzold GL, Swenberg JA. 1978. Detection of DNA damage induced in vivo following exposure of rats to carcinogens. Cancer Res 38:1589-1594. *Phillips JC, Lake BC, Heading CE, et a1. 1975. Studies on the metabolism of dimethylnitrosamine in the rat: I. Effect of dose, route of administration and sex. Fd Cosmet Toxicol 132203-209 *Pignatelli B, Richard I, Bourgade M, et a1. 1987. An improved method for analysis of total N-nitroso compounds in gastric juice In: Relevance of N-nitroso compounds to Human Cancer: Exposures and Mechanisms. IARC Sci Publ 84:209-215.. *Poirier S, G Hubert, de-The H et a1. 1987. Occurrence of volatile nitrosamines in food samples collected in three high-risk areas for nasopharyngeal carcinoma. IARC Sci Publ 84:415-419. *Polo J, Chow UL. 1976. Efficient photolytic degradation of nitrosamines. J Natl Cancer Inst 56:997—1001. 108 8. REFERENCES *Prasanna HR, Lotlikar PD, Barndt C, et a1. 1985. Factors influencing the activity of dimethylnitrosamine demethylase in hamster, rat and chicken liver microsomes. Cancer Lett 26:25-32. *Preussmann R. 1984. Occurrence and exposure to N-nitroso compounds and precursors. IARC Sci Publ. 57:3-15. *Prival MJ, Mitchell VD. 1981. Influence of microsomal and cytosolic fractions from rat, mouse, and hamster liver on the mutagenicity of dimethylnitrosamine in the Salmonella plate incorporation assay. Cancer Res 41:4361-4367. *Pylypiw HM Jr. 1987. The development and application of analytical methods for the study of volatile and non-volatile N-nitrosamines (Pharmacokinetics). Disser Abstr Inter 48:1028-B. *Pylypiw HM Jr., Zimmerman F, Harrington CW, et a1. 1985. Apparatus for trace determination of volatile N-nitrosamines in small samples. Anal Chem 57:2996-2997. *Raabe DC. 1986. Inhalation uptake of selected chemical vapors at trace levels. Report to California state Air Resources Board, Sacramento. NTIS PB86-209863. *Rao GS, Osborn JC, Adatia MR. 1982. Drug-nitrite interactions in human saliva: effects of food constituents on carcinogenic N-nitrosamine formation. J Dent Res 61:768-771. *Reuber MD. 1975. Hepatic vein thrombosis in Buffalo strain female rats ingesting dimethylnitrosamine. Pathol Europ 10:241-244. *Rhoades JW, Hosenfeld JM, Taylor JM, et a1. 1980. Comparison of analysis of wastewaters for N-nitrosamines using various detectors. IARC Sci Publ 31:377-381. *Ross RD, Morrison J, Rounbehler DP, et a1. 1977. N-Nitroso compound impurities in herbicide formulations. J Agric Food Chem 25:1416-1418. *Rounbehler DP, Reisch J, Fine DH. 1980. Nitrosamines in new motor-cars. Fd Cosmet Toxicol 18:147-151. *RTECS. 1988. Registry of Toxic Effects of Chemical Substances. On-line: 08/05/88. Washington, DC: National Institute for Occupational Safety and Health. *Sabljic A. 1987. On the prediction of soil sorption coefficients of organic pollutants from molecular structure: application of molecular topology model. Environ. Sci Technol 21:358-366. 109 8. REFERENCES *Sakai A, Inoue T, Tanimura A. 1984. Formation of Volatile Nitrosamines by Drug~Nitrite Interactions Under Physiological Conditions. Gann 75:245-252. *SANSS, 1988. Structure and Nomenclature Search System. On-line: 07/28/88. Chemical Tnformation System. *Sawicki E, Belsky T, Friedel RA, et al. 1977. N—nitrosodimethylamine in ambient air. Analytical method. Health Lab Sci 14:59—63. *Scanlan RA, Barbour JF, Hotchkiss JH, et al. 1980. N-Nitrosodimethylamine in beer. Fd Cosmet Toxicol 18:27-30. *Scanlan RA. 1983. Formation and Occurrence of Nitrosamines in Food. Cancer Res. (Suppl.) 43:24355-24403. *Scherf HR, Schmahl D. 1975. Experimental investigations on immuno- depressive properties of carcinogenic substances in male Sprague-Dawley rats. 52:76-87. *Sen NP, Seaman S. 1981a. Gas liquid chromatographic-thermal energy analyzer determination of N-nitrosodimethylamine in beer at low parts per billion level. J Assoc Off Anal Chem 64:933-938. *Sen NP, Seaman S. 1981b. Possible presence of volatile N-nitrosamines in cooking oils, margarine and butter. J Agric Food Chem 29:787-789. *Sen NP, Seaman S, Karpinsky K. 1984. Determination of N-nitrosodimethylamine in nonfat dry milk: collaborative study. J Assoc Off Anal Chem 67:232-236. *Sen NP, Tessier L, Seaman SW, et al. 1985a. Volatile and nonvolatile nitrosamines in fish and the effect of deliberate nitrosation under simulated gastric conditions. J Agric Food Chem 33:264-268. *Sen NP, Kushwaha SC, Seaman SW, et al. 1985b. Nitrosamines in baby bottle nipples and pacifiers: occurrence, migration and effect of infant formulas and fruit juices on in vitro formation of nitrosamines under simulated gastric conditions. J Agric Food Chem 33:428-433. *Shapley D. 1976. Nitrosamines: scientists on the trail of prime suspect in urban cancer. Science 1912268-270. ‘ *Sharma RK, Lemmon M, Bakke J, et al. 1983. Studies of in utero sister chromatid exchange induction and cell replication kinetics [abstract]. Environ Mutagen 5:406. *Sharma RK, Jacobson—Kram D, Lemmon M et a1. 1985. Sister chromatid and cell replication kinetics in fetal and material cells after treatment with chemical teratogens. Mutat Res 158:217-231. 110 8. REFERENCES *Shimizu RW, Sun JD, Li AP, et a1. 1984. The use of sister-chromatid exchange in Chinese hamster primary lung cell cultures to measure genotoxicity. Mutat Res 130:333-342. *Singer B. 1979. N-nitroso alkylating agents: formation and persistence of alkyl derivatives in mammalian as contributing factors in carcinogens. J Natl Cancer Inst 62:1329-1339. *Sirianni SR, Huang CC. 1987. Comparison of S9 fractions from rats, mice, and Chinese hamsters to metabolize dimethylnitrosamine and diethylnitrosamine to intermediates that induce sister-chromatid exchanges in V79 cells. Mutat Res 188:7-11. *Skaare JU, Nafstad I; 1978. Interaction of vitamin E and selenium with the hepatotoxic agent dimethylnitrosamine. Acta Pharmacol Toxicol 43;119-128. *Song PJ, Hu JF. 1988. N-Nitrosamines in Chinese foods. Fd Chem Toxicol 26:205—208. *Spiegelhalder B. 1984. Occupational exposure to N-nitrosamines. Air measurements and biological monitoring. IARC Sci Publ 57:937-942. *Spiegelhalder B, Preussmann R. 1984. Contamination of toiletries and cosmetic products with volatile and nonvolatile N-nitroso carcinogens. J Cancer Res Clin Oncol 1082160-163. *Spiegelhalder B, Preussmann R. 1985. In vivo nitrosation of amidopyrine in humans: use of ‘ethanol effect' for biological monitoring of N-nitrosodimethylamine in urine. Carcinogenesis 6:545-548. *Spiegelhalder B, Eisenbrand G, Preussmann R. 1979. Contamination of beer with trace quantities of N-nitrosodimethylamine. Fd Cosmet Toxicol 17:29-31. *Spiegelhalder B, Eisenbrand G, Preussmann R. 1982. Urinary excretion of N-nitrosamines in rats and humans. IARC Sci Publ 41:443-449. *State of Kentucky. 1986. New or Modified Sources Emitting Toxic Air Pollution. 401 KAR 63:022. *Steinmetz KL, Mirsalis JC. 1984. Induction of unscheduled DNA synthesis in primary cultures of rat pancreatic cells following in vivo and in vitro treatment with genotoxic agents. Environ Mutagen 6:321-330. *Strombeck DR, Harrold D, Rogers Q, et al. 1983. Plasma amino acid, glucagon, and insulin concentrations in dogs with nitrosamine-induced hepatic disease. Am J Vet Res 44:2028-2036. 111 8. REFERENCES *Stumpf R, Margison GP, Montesano R, Pegg AE. 1979. Formation and loss of alkylated purines from DNA of hamster liver after administration of dimethylnitrosamine. Cancer Res 39:50-54. *Sumi Y, Miyakawa M. 1983. Susceptibility of germ-free rats to the hepatotoxic effects of dimethylnitrosamine or dimethylamine plus sodium nitrite administered orally. Cancer Res 43:2942-2946. *Swann RL, Laskowski DA, McCall PJ, et a1. 1983. A rapid Method for the Estimation of the Environmental Parameters Octanol/Water Partition Coefficient, Soil Sorption Constant, Water to Air Ratio and Water Solubility. Res Rev 85:17-28. *Swann PF. 1984. Effect of ethanol on nitrosamine metabolism and distribution. Implications for the role of nitrosamines in human cancer and for the influence of alcohol consumption on cancer incidence. IARC Sci Publ 57:501-512. *Swann PF, Coe AM, Mace R. 1984. Ethanol and dimethylnitrosamine and diethylnitrosamine metabolism and disposition in the rat. Possible relevance to the influence of ethanol on human cancer incidence. Carcinogenesis 5:1337-1343. *Tabak HH, Quave SA, Mashni CI, et a1. 1981A. Biodegradability studies for predicting the environmental fate of organic priority pollutants. In: Test protocols for environmental fate and movement of toxicants. Proceedings of 94th annual meeting of symposium association of official Anal Chem, Washington, DC, 267—327. *Takayama S, Oota K. 1965. Induction of malignant tumors in various strains of mice by oral administration of N-nitrosodimethylamine and N-nitrosodiethylamine. Gann 56:189-199. *Tate RL III, Alexander M. 1975. Stability of nitrosamines in samples of lake water, soil, and sewage. J Natl Cancer Inst. 54:327—330. *Tates AD, Neuteboom I, Hofker M., et a1. 1980. A micronucleus technique for detecting clastogenic effects of mutagens/carcinogens (DEN, DMN) in hepatocytes of rat liver in vivo. Mutat Res 74:11-20. *Telling GM. 1972. A gas-liquid chromatographic procedure for the detection of volatile N-nitrosamines at the ten parts per billion level in foodstuffs after conversion to their corresponding nitramines. J Chromatog 73:79-87. *Terao K, Aikawa T, Kera K. 1978. A synergistic effect of nitrosodimethylamine on sterigmatocystin carcinogenesis in rats. Fd Cosmet Toxicol 16:591-596. 112 8. REFERENCES *Terracini B, Palestro G, Ramella GM, et al. 1966. Carcinogenicity of dimethylnitrosamine in Swiss mice. Br J Cancer 20:871-876. *Terracini B, Magee PN, Barnes JM. 1967. Hepatic pathology in rats on low dietary levels of dimethylnitrosamine. Br J Cancer 21:559-565. *Thomas RG. 1982. Volatilization from water. In: Lyman WJ, Reehl WF, Rosenblatt DH, eds. Handbook of chemical property estimation methods. New York, NY: McGraw-Hill Book Co., 15-27. *Thomas P, Fugmann R, Aranyi C, et al. 1985. The effect of dimethylnitrosamine on host resistance and immunity. Toxicol Appl Pharmacol 77:219-229. *Thompson HC Jr, Billedeau SM, Miller BJ. 1986. Gas chromatographic thermal energy analysis method for N-nitrosodibutylamine in latex infant pacifiers: collaborative study. J Assoc Off Anal Chem 69:504-507. *Tomkins DJ, Kwok SE, Douglas GR, et a1. 1982. Sister chromatid exchange response of human diploid fibroblasts and Chinese hamster ovary cells to dimethylnitrosamine and benzo(a)pyrene. Environ Mutagen 4:203-214. *Trzos RJ, Petzold GL, Brunden MN, et a1. 1978. The evaluation of sixteen carcinogens in the rat using the micronucleus test. Mutat Res 58:79-86. *Tuazon EC, Carter WPL, Atkinson R, et a1. 1984. Atmospheric reactions of N-nitrosodimethylamine and dimethylnitramine. Environ Sci Tech 18:49-54. *Ungar H. 1984. Primary portal venopathy in the golden hamster treated with low doses of dimethylnitrosamine. Liver 4:244-254. *Ungar H. 1986. Venoocclusive disease of the liver and phlebectatic peliosis in the golden hamster exposed to dimethylnitrosamine. Path Res Pract 181:180-187. *Usero JL, Del Arco A, Izquierdo C, et a1. 1987. Comparative study of high- performance liquid chromatography with ultraviolet detection and gas chromatography with the Hall electrolytic conductivity detector in the analysis of N-nitroso compounds. J Chromatog 407:355-362. VIAR. 1987. Contract Laboratory Program Statistical Database. Output date: April 13, 1987. *VIEW Database. 1989. Agency for Toxic Substances and Disease Registry (ATSDR), Office of External Affairs, Exposure and Disease Registry Branch, Atlanta, CA. June, 1989. 113 8. REFERENCES » *Wade GG, Mandel R, Ryser HJ-P. 1987. Marked synergism of dimethylnitrosamine carcinogenesis in rats exposed to cadmium. Cancer Res 47:6606-6613. *Walker EA, Castegnaro M, Garren L, et a1. 1978. Limitations to the protective effect of rubber gloves for handling nitrosamines. IARC Sci Publ 19:535-543. *Waynforth HB, Magee PN. 1974. The effect of N-nitroso-N-methylurea and N-dimethylnitrosamine on cell mediated and humoral immune responses in rats and mice. Br J Cancer 30:512-523. *Weast RC, ed. 1983. CRC handbook of chemistry and physics. 64th ed. Boca Raton, FL: CRC Press, C-270. *Webb KS, Cough TA, Garrick A, et a1. 1979. Mass spectrometric and chemiluminescent detection of picogram amounts of N-nitrosodimethylamine. Anal Chem 51;989-992. *Weston RJ. 1984. Analysis of cereals, malted foods and dried legumes for N-nitrosodimethylamine. J Sci Food Agric 35:782-786. *Wild CP, Lu SH, Montesano R. 1987. Radioimmunoassay used to detect DNA alkylation adducts in tissues from populations at high risk for oesophageal and stomach cancer. Iarc Sci Publ 84:534-537. *Windholz M, ed. 1983. The Merck index. 10th ed. Rahway, NJ: Merck and Co., Inc., 952, 864, 1387—1388. *Wishnok JS, Rogers AE, Sanchez 0, et a1. 1978. Dietary effects on the pharmacokinetics of three carcinogenic nitrosamines. Toxicol Appl Pharmacol 43:391-398. *Wyrobek AJ, Bruce WR. 1975. Chemical induction of sperm abnormalities in mice. Proc Nat Acad Sci. 72:4425-4429. *Xesarone CF, Bolognesi C, Santi L. 1979. DNA repair synthesis in mice spermatids after treatment with N-methyl-N-nitroso-urea and N-dimethyl- nitrosamine: preliminary results. Toxicology 12:183-186. *Yamane Y, Sakai K, Umeda T, et a1. 1984. Suppressive effect of cupric acetate on DNA alkylation, DNA synthesis and tumorigenesis in the liver of dimethylnitrosamine-treated rats. Gann 75:1062—1069. *Yoo J-SH, Guengerich FP, Yang CS. 1988. Metabolism of N-nitrosodialkylamines by human liver microsomes. Cancer Res 88:1499-1505. *Yu MC, Henderson BE. 1987. Intake of Cantonese-style salted fish as a cause of nasopharyngeal carcinoma. IARC Sci Publ 84:547-549. 114 8. REFERENCES *Zeisel SH, daCosta KA, LaMont JT. 1988. Mono-, di- and trimethylamine in human gastric fluid: Potential substrates for nitrosodimethylamine formation. Carcinogenesis 9:179-181. *Zoeteman BCJ, Harmsen K, Linders JBHJ, et a1. 1980. Persistent organic pollutants in river water and ground water of The Netherlands. Chemosphere 9:231-249. 115 9. GLOSSARY Acute Exposure -- Exposure to a chemical for a duration of 14 days or less, as specified in the Toxicological Profiles. Adsorption Coefficient (Koo) -- The ratio of the amount of a chemical adsorbed per unit weight of organic carbon in the soil or sediment to the concentration of the chemical in solution at equilibrium. Adsorption Ratio (Kd) -- The amount of a chemical adsorbed by a sediment or soil (i.e., the solid phase) divided by the amount of chemical in the solution phase, which is in equilibrium with the solid phase, at a fixed solid/solution ratio. It is generally expressed in micrograms of chemical sorbed per gram of soil or sediment. Bioconcentration Factor (BCF) -- The quotient of the concentration of a chemical in aquatic organisms at a specific time or during a discrete time period of exposure divided by the concentration in the surrounding water at the same time or during the same time period. Cancer Effect Level (CEL) -- The lowest dose of chemical in a study or group of studies which produces significant increases in incidence of cancer (or tumors) between the exposed populaton and its appropriate control. Carcinogen -- A chemical capable of inducing cancer. Ceiling Value (CL) -- A concentration of a substance that should not be exceeded, even instantaneously. Chronic Exposure -- Exposure to a chemical for 365 days or more, as specified in the Toxicological Profiles. Developmental Toxicity -- The occurrence of adverse effects on the developing organism that may result from exposure to a chemical prior to conception (either parent), during prenatal development, or postnatally to the time of sexual maturation. Adverse developmental effects may be detected at any point in the life span of the organism. Embryotoxicity and Fetotoxicity -- Any toxic effect on the conceptus as a result of prenatal exposure to a chemical; the distinguishing feature between the two terms is the stage of development during which the insult occurred. The terms, as used here, include malformations and variations, altered growth, and in utero death. EPA Health Advisory -- An estimate of acceptable drinking water levels for a chemical substance based on health effects information. A health advisory is not a legally enforceable federal standard, but serves as technical guidance to assist federal, state, and local officials. 116 9. GLOSSARY Immediately Dangerous to Life or Health (IDIH) -- The maximum environmental concentration of a contaminant from which one could escape within 30 min without any escape-impairing symptoms or irreversible health effects. Intermediate Exposure -- Exposure to a chemical for a duration of 15—364 days, as specified in the Toxicological Profiles. Immunologic Toxicity -- The occurrence of adverse effects on the immune system that may result from exposure to environmental agents such as chemicals. In vitro -- Isolated from the living organism and artificially maintained, as in a test tube. In vivo -- Occurring within the living organism. Lethal Concentration(ID) (L610) -- The lowest concentration of a chemical in air which has been reported to have caused death in humans or animals. Lethal Concentration(50) (L650) —- A calculated concentration of a chemical in air to which exposure for a specific length of time is expected to cause death in 50% of a defined experimental animal population. Lethal Dose(lD) (LDLD) -- The lowest dose of a chemical introduced by a route other than inhalation that is expected to have caused death in humans or animals. Lethal Dose(50) (LD50) -- The dose of a chemical which has been calculated to cause death in 50% of a defined experimental animal population. Lowest—Observed-Adverse-Effect Level (LOAEL) -- The lowest dose of chemical in a study or group of studies which produces statistically or biologically significant increases in frequency or severity of adverse effects between the exposed population and its appropriate control. LT50 (lethal time) -- A calculated period of time within which a specific concentration of a chemical is expected to cause death in 50% of a defined experimental animal population. Malformations -- Permanent structural changes that may adversely affect survival, development, or function. Minimal Risk Level -- An estimate of daily human exposure to a chemical that is likely to be without an appreciable risk of deleterious effects (noncancerous) over a specified duration of exposure. 117 9. GLOSSARY Mutagen -- A substance that causes mutations. A mutation is a change in the genetic material in a body cell. Mutations can lead to birth defects, miscarriages, or cancer. Neurotoxicity -- The occurrence of adverse effects on the nervous system following exposure to a chemical. No—Observed—Adverse-Effect Level (NOAEL) -- That dose of chemical at which there are no statistically or biologically significant increases in frequency or severity of adverse effects seen between the exposed population and its appropriate control. Effects may be produced at this dose, but they are not considered to be adverse. Octanol-Water Partition Coefficient (Row) -- The equilibrium ratio of the concentrations of a chemical in n-octanol and water, in dilute solution. Permissible Exposure Limit (PEL) -- An allowable exposure level in workplace air averaged over an 8—h shift. q1* -- The upper-bound estimate of the low-dose slope of the dose-response curve as determined by the multistage procedure. The ql* can be used to calculate an estimate of carcinogenic potency, the incremental excess cancer risk per unit of exposure (usually g/L for water, mg/kg/day for food, and g/m3 for air). Reference Dose (RfD) -- An estimate (with uncertainty spanning perhaps an order of magnitude) of the daily exposure of the human population to a potential hazard that is likely to be without risk of deleterious effects during a lifetime. The RfD is operationally derived from the NOAEL (from animal and human studies) by a consistent application of uncertainty factors that reflect various types of data used to estimate Rst and an additional modifying factor, which is based on a professional judgment of the entire database on the chemical. The Rst are not applicable to nonthreshold effects such as cancer. Reportable Quantity (RQ) -- The quantity of a hazardous substance that is considered reportable under CERCLA. Reportable quantities are: (l) 1 lb or greater or (2) for selected substances, an amount established by regulation either under CERCLA or under Sect. 311 of the Clean Water Act. Quantities are measured over a 24-h period. Reproductive Toxicity -- The occurrence of adverse effects on the reproductive system that may result from exposure to a chemical. The toxicity may be directed to the reproductive organs and/or the related. endocrine system. The manifestation of such toxicity may be noted as alterations in sexual behavior, fertility, pregnancy outcomes, or modifications in other functions that are dependent on the integrity of this system. 118 9. GLOSSARY Short—Term Exposure Limit (STEL) -- The maximum concentration to which workers can be exposed for up to 15 min continually. No more than four excursions are allowed per day, and there must be at least 60 min between exposure periods. The daily TLV-TWA may not be exceeded. Target Organ Toxicity -- This term covers a broad range of adverse effects on target organs or physiological systems (e.g., renal, cardiovascular) extending from those arising through a single limited exposure to those assumed over a lifetime of exposure to a chemical. TD50 (toxic dose) -- A calculated dose of a chemical, introduced by a route other than inhalation, which is expected to cause a specific toxic effect in 50% of a defined experimental animal population. Teratogen -- A chemical that causes structural defects that affect the development of an organism. Threshold Limit Value (TLV) -- A concentration of a substance to which most workers can be exposed without adverse effect. The TLV may be expressed as a TWA, as a STEL, or as a CL. Time-weighted Average (TWA) -- An allowable exposure concentration averaged over a normal 8-h workday or 40-h workweek. Uncertainty Factor (UF) -- A factor used in operationally deriving the RfD from experimental data. UFs are intended to account for (l) the variation in sensitivity among the members of the human population, (2) the uncertainty in extrapolating animal data to the case of humans, (3) the uncertainty in extrapolating from data obtained in a study that is of less than lifetime exposure, and (A) the uncertainty in using LOAEL data rather than NOAEL data. Usually each of these factors is set equal to 10. 119 APPENDIX: PEER REVIEW A peer review panel was assembled for N—nitrosodimethylamine. The panel consisted of the following members: Dr. Russell Cattley, Department of Microbiology, Pathology and Parasitology, College of Veterinary Medicine, North Carolina State University; Dr. Elaine Faustman, Department of Environmental Health, University of Washington; Dr. James Felton, Molecular Biology Section, Lawrence Livermore National Laboratory, University of California; Dr. Freddy Hamburger, Bio-Research Consultants, Inc; and Dr. Raymond Smith, Department of Pathology and Microbiology, University of Nebraska Medical Center. These experts collectively have knowledge of N- nitrosodimethylamine's physical and chemical properties, toxicokinetics, key health end points, mechanisms of action, human and animal exposure, and quantification of risk to humans. All reviewers were selected in conformity with the conditions for peer review specified in the Superfund Amendments and Reauthorization Act of 1986, Section 110. A joint panel of scientists from ATSDR and EPA has reviewed the peer reviewers' comments and determined which comments will be included in the profile. A listing of the peer reviewers' comments not incorporated in the profile, with a brief explanation of the rationale for their exclusion, exists as part of the administrative record for this compound. A list of databases reviewed and a list of unpublished documents cited are also included in the administrative record. The citation of the peer review panel should not be understood to imply their approval of the profile's final content. The responsibility for the content of this profile lies with the Agency for Toxic Substances and Disease Registry. aU.S.GOVERNMENTPRlNTINGOFHCE:199 o -73 2 -3 3 2/ C. BERKELEY LIBRARIES lilfllllllllllfilllfllll (0555557'47 DEPARTMENT OF HEALTH 8| HUMAN SERVICES Public Health Service Centers for Disease Control Atlanta,GA 30333 SPECIAL FOURTH-CLASS RATE POSTAGE a. FEES PAID PHS/CDC Permit No. G-284 Official Business Penalty for Ptivate Use $300