United States Environmental Protection 1=1 m m Agency EPA/690/R-07/020F Final 5-30-2007 Provisional Peer Reviewed Toxicity Values for Stable Lutetium (CASRN 7439-94-3) Superfund Health Risk Technical Support Center National Center for Environmental Assessment Office of Research and Development U.S. Environmental Protection Agency Cincinnati, OH 45268 ------- Acronyms and Abbreviations bw body weight cc cubic centimeters CD Caesarean Delivered CERCLA Comprehensive Environmental Response, Compensation and Liability Act of 1980 CNS central nervous system cu.m cubic meter DWEL Drinking Water Equivalent Level FEL frank-effect level FIFRA Federal Insecticide, Fungicide, and Rodenticide Act g grams GI gastrointestinal HEC human equivalent concentration Hgb hemoglobin i.m. intramuscular i.p. intraperitoneal IRIS Integrated Risk Information System IUR inhalation unit risk i.v. intravenous kg kilogram L liter LEL lowest-effect level LOAEL lowest-observed-adverse-effect level LOAEL(ADJ) LOAEL adjusted to continuous exposure duration LOAEL(HEC) LOAEL adjusted for dosimetric differences across species to a human m meter MCL maximum contaminant level MCLG maximum contaminant level goal MF modifying factor mg milligram mg/kg milligrams per kilogram mg/L milligrams per liter MRL minimal risk level MTD maximum tolerated dose MTL median threshold limit NAAQS National Ambient Air Quality Standards NOAEL no-ob served-adverse-effect level NOAEL(ADJ) NOAEL adjusted to continuous exposure duration NOAEL(HEC) NOAEL adjusted for dosimetric differences across species to a human NOEL no-ob served-effect level OSF oral slope factor p-IUR provisional inhalation unit risk p-OSF provisional oral slope factor p-RfC provisional inhalation reference concentration 1 ------- p-RfD provisional oral reference dose PBPK physiologically based pharmacokinetic ppb parts per billion ppm parts per million PPRTV Provisional Peer Reviewed Toxicity Value RBC red blood cell(s) RCRA Resource Conservation and Recovery Act RDDR Regional deposited dose ratio (for the indicated lung region) REL relative exposure level RfC inhalation reference concentration RfD oral reference dose RGDR Regional gas dose ratio (for the indicated lung region) s.c. subcutaneous SCE sister chromatid exchange SDWA Safe Drinking Water Act sq.cm. square centimeters TSCA Toxic Substances Control Act UF uncertainty factor l^g microgram [j,mol micromoles voc volatile organic compound 11 ------- 5-30-2007 PROVISIONAL PEER REVIEWED TOXICITY VALUES FOR STABLE LUTETIUM (CASRN 7439-94-3) Background On December 5, 2003, the U.S. Environmental Protection Agency's (EPA's) Office of Superfund Remediation and Technology Innovation (OSRTI) revised its hierarchy of human health toxicity values for Superfund risk assessments, establishing the following three tiers as the new hierarchy: 1. EPA's Integrated Risk Information System (IRIS). 2. Provisional Peer-Reviewed Toxicity Values (PPRTV) used in EPA's Superfund Program. 3. Other (peer-reviewed) toxicity values, including: ~ Minimal Risk Levels produced by the Agency for Toxic Substances and Disease Registry (ATSDR), ~ California Environmental Protection Agency (CalEPA) values, and ~ EPA Health Effects Assessment Summary Table (HEAST) values. A PPRTV is defined as a toxicity value derived for use in the Superfund Program when such a value is not available in EPA's Integrated Risk Information System (IRIS). PPRTVs are developed according to a Standard Operating Procedure (SOP) and are derived after a review of the relevant scientific literature using the same methods, sources of data, and Agency guidance for value derivation generally used by the EPA IRIS Program. All provisional toxicity values receive internal review by two EPA scientists and external peer review by three independently selected scientific experts. PPRTVs differ from IRIS values in that PPRTVs do not receive the multi-program consensus review provided for IRIS values. This is because IRIS values are generally intended to be used in all EPA programs, while PPRTVs are developed specifically for the Superfund Program. Because new information becomes available and scientific methods improve over time, PPRTVs are reviewed on a five-year basis and updated into the active database. Once an IRIS value for a specific chemical becomes available for Agency review, the analogous PPRTV for that same chemical is retired. It should also be noted that some PPRTV manuscripts conclude that a PPRTV cannot be derived based on inadequate data. Disclaimers Users of this document should first check to see if any IRIS values exist for the chemical of concern before proceeding to use a PPRTV. If no IRIS value is available, staff in the regional Superfund and RCRA program offices are advised to carefully review the information provided in this document to ensure that the PPRTVs used are appropriate for the types of exposures and circumstances at the Superfund site or RCRA facility in question. PPRTVs are periodically updated; therefore, users should ensure that the values contained in the PPRTV are current at the time of use. 3 ------- 5-30-2007 It is important to remember that a provisional value alone tells very little about the adverse effects of a chemical or the quality of evidence on which the value is based. Therefore, users are strongly encouraged to read the entire PPRTV manuscript and understand the strengths and limitations of the derived provisional values. PPRTVs are developed by the EPA Office of Research and Development's National Center for Environmental Assessment, Superfund Health Risk Technical Support Center for OSRTI. Other EPA programs or external parties who may choose of their own initiative to use these PPRTVs are advised that Superfund resources will not generally be used to respond to challenges of PPRTVs used in a context outside of the Superfund Program. Questions Regarding PPRTVs Questions regarding the contents of the PPRTVs and their appropriate use (e.g., on chemicals not covered, or whether chemicals have pending IRIS toxicity values) may be directed to the EPA Office of Research and Development's National Center for Environmental Assessment, Superfund Health Risk Technical Support Center (513-569-7300), or OSRTI. INTRODUCTION An assessment for stable, nonradioactive lutetium is not available on IRIS (U.S. EPA, 2007), the HEAST (U.S. EPA, 1997), or the Drinking Water Standards and Health Advisories list (U.S. EPA, 2004). No relevant documents were located in the Chemical Assessment and Related Activities (CARA) list (U.S. EPA, 1991, 1994). AT SDR (2006), NTP (2006), IARC (2006) and WHO (2006) have not produced documents regarding lutetium. Comprehensive literature searches were conducted in 1998 of the following databases: TOXLINE (1965-1998), CANCERLINE (1970-1998), MEDLINE (1966-1998), GENETOX, DART, CCRIS, CHEMID, RTECS, EMIC, ETICBACK and TSCATS for toxicity studies of lutetium. Update literature searches were conducted from 1998 through June 2003 in: TOXLINE (supplemented with BIOSIS and NTIS updates), CANCERLIT, MEDLINE, CCRIS, GENETOX, HSDB, DART/ETICBACK, EMIC/EMICBACK, RTECS and TSCATS. An update literature search covering 2003 through November 2005 was conducted in the following databases: MEDLINE, TOXLINE SPECIAL, TOXCENTER BIOSIS, TSCATS, CCRIS, DART/ETIC, GENETOX, HSDB, RTECS and CURRENT CONTENTS. Searches of Medline and Current Contents from November 2005 to April 2006 were also conducted. REVIEW OF PERTINENT DATA Human Studies No data regarding the toxicity of lutetium to humans following oral exposure were located. Lutetium Texaphyrin (Lu-Tex) has been used in the treatment of age-related macular degeneration. The reported effective dose range was 2 to 4 mg/kg-day of Lu-Tex (Pharmacyclic, 1999). The Texaphyrin moiety is a large porphyrin structure with a number of side chains and is probably at least as important as lutetium, itself, for its biological action. Therefore, any dose- 4 ------- 5-30-2007 response information for the compound cannot be applied to the assessment of lutetium, alone. Details of the responses encountered in clinical trials were not reported. In addition, even an approximate molecular weight for Lu-Tex cannot be estimated, such that the dose range for lutetium cannot be determined. No data regarding the toxicity of lutetium to humans following inhalation exposure were located. The pulmonary toxicity of inhaled rare earth compounds, in general, has been the subject of debate, especially with regard to the relative contributions of radioactive contaminants versus stable elements in the development of progressive pulmonary interstitial fibrosis (Haley, 1991; Beliles, 1994). In particular, although it is known that stable rare earths compounds can produce a static, foreign-body-type lesion consistent with benign pneumoconiosis, there is uncertainty whether they can also induce interstitial fibrosis that progresses after termination of exposure. Human inhalation toxicity data on stable rare earth elements mainly consist of case reports on workers exposed to multiple lanthanides (Sulotto et al., 1986; Kappenberger and Buhlmann, 1975; Husain et al., 1980; Sabbioni et al., 1982; Vocaturo et al., 1983; Colombo et al., 1983; Vogt et al., 1986; Waring and Watling, 1990; Deng et al., 1991). No data regarding the carcinogenicity of stable lutetium in humans were located. Animal Studies Groups of six male and six female CRW rats were fed 0, 0.01, 0.1 or 1% lutetium chloride (purity not reported) in the diet for 90 days (Haley et al., 1964). Food intake was not reported. Compound intake, as lutetium chloride (trichloride), was estimated to have been 8.1, 81 or 810 mg/kg-day in males, and 9.5, 95 or 950 mg/kg-day in females, using body weight (growth curve) data reported in the study and food consumption estimates based on allometric scaling (U.S. EPA, 1988). Based on the weight-fraction of lutetium in lutetium chloride (0.622), the corresponding lutetium intakes were calculated to be 5.0, 50 and 504 mg/kg-day for males and 5.9, 59 or 590 mg/kg-day for females. Body weight and hematology (total erythrocytes, total leukocytes, differential cell count, platelets, hemoglobin and hematocrit) were measured biweekly, and gross and histological examinations (heart, lung, liver, kidney, pancreas, spleen, adrenal and small intestine) were performed at the end of the study. No exposure-related changes in these endpoints were observed in either sex. No adverse effects were identified in this study, making the high dose of 504-590 mg Lu/kg-day a freestanding NOAEL. Animal inhalation toxicity data on stable rare earths mainly consist of a few inhalation or intratracheal studies on some rare earth mixtures and some single compounds (Schepers, 1955a, 1955b; Schepers et al., 1955; Ball and Van Gelder, 1966; Abel and Talbot, 1967; Mogilevskaya and Raikhlin, 1967). No lutetium-specific data were found, however. A comprehensive assessment of the human and animal data by Haley (1991) concluded that the evidence suggests that inhalation exposure to high concentrations of stable rare earths can produce lesions compatible with pneumoconiosis and progressive pulmonary fibrosis, and that the potential for inducing these lesions is related to chemical type, physiochemical form, and dose and duration of exposure. 5 ------- 5-30-2007 No data regarding the carcinogenicity of stable lutetium in animals were located in the available literature. No histological changes were found in rats that were orally-exposed to lutetium chloride for 90 days (Haley et al., 1964), but this study is inadequate for assessing carcinogenic effects due to insufficient duration of exposure and lack of a post-treatment observation period. Other Studies Acute oral and intraperitoneal LD50 values of 4416 and 196 mg Lu/kg, respectively, were determined for lutetium chloride in 50 male CF1 mice observed for 7 days (Haley et al., 1964). Symptoms of acute lutetium chloride toxicity included ataxia, writhing, labored respiration, walking on toes with back arched and sedation (Haley et al., 1964). The peak death rate was reached at 48 hours after exposure, but some deaths occurred at 24 hours. Acute intraperitoneal LD50 values of 108 and 125 mg Lu/kg were determined for lutetium nitrate in female mice and rats, respectively, observed for 30 days (Bruce et al., 1963). Graca et al. (1962) reported an LD50 of 135 mg/kg for lutetium citrate (84 mg Lu/kg) administered intraperitoneally in mice observed for 168 hours, and an LD50 of 81 mg/kg for lutetium citrate (50 mg Lu/kg) in guinea pigs treated similarly. Durbin et al. (1956) evaluated the metabolism and elimination of radioisotopes of 15 lanthanide elements in groups of 5 female Sprague-Dawley rats (including 177Lu given in intramuscular doses of 7.3 to 29 ju.Ci). Animals treated with 177Lu were sacrificed after 1, 4 and 16 days. Urinary and fecal excreta were collected daily for the first 4 days and twice weekly thereafter; upon necropsy, radioactivity was measured in various tissues. Although details of the method used to estimate absorption were not provided, the authors reported that gastrointestinal absorption of gavage doses of 144Ce, 152'154Eu, 160Tb and 170Tm was less than 0.1% of the administered dose, suggesting that oral absorption of lutetium is likely to be low. Based on graphical presentation of the results, approximately 20% of the absorbed dose of 177Lu was excreted, 65% deposited in bone and less than 5% deposited in liver (remaining 10% not quantified by tissue) 1 day after injection. Although long-term skeletal retention of 177Lu was not evaluated in the study, the authors reported that the half-life for elimination from the skeleton of the heavier lanthanides (lutetium is a heavy lanthanide) was about 2.5 years, based on data collected for 160Tb and 170Tm over 256 days. Graca et al. (1964) investigated the effects of intravenously-administered compounds (chlorides, citrates and edetates) of rare-earth elements on heart rate, blood pressure, respiration and clinical hematology in male and female dogs (breed and number/sex not specified). Aqueous solutions of the compounds (equivalent to 5% of the chloride) of 15 rare earth elements were injected into a cannula inserted into the left femoral vein. Ten doses of 10 mg/kg (as chloride) each were injected under anesthesia at 10-minute intervals. For each rare-earth element, nine dogs were divided into groups of three each treated with the chloride, citrate and edetate. Three groups of control dogs were injected with sodium citrate (n=6), ammonium versenate (n=6) or Ringer's solution (n=12) in the same manner as treated animals. Blood samples were collected from the right femoral vein before treatment and 0, 10, 30, 60, 100 and 160 minutes after treatment for analysis of erythrocyte, leukocyte, and differential cell counts, prothrombin and coagulation time, hemoglobin, sedimentation and hematocrit. After 6 ------- 5-30-2007 160 minutes, the animals were necrospied and tissues were collected for histopathology (liver, spleen, kidney, lung, sternum, mesentery lymph nodes, heart, adrenal and ovaries or testes). Heart rate, respiration and blood pressure readings were made at the same intervals as blood samples. Results for the 15 elements were discussed generally and presented graphically as change over time after treatment (Graca et al., 1964). Some animals died from treatment (14/45 treated with chlorides, 4/45 treated with citrates, and 1/45 treated with edetates), but the mortality was not reported by element. In general, the chloride compounds were more toxic than the citrate or edetate compounds. Lutetium chloride treatment resulted in a transient spike in blood pressure (150% of pretreatment values) and heart rate (about 130% of pretreatment values) at 60 minutes post-treatment; results for the citrate and edetate appeared to be within the range of control values, based on the graphs. Respiratory rates appeared to be within control values for all lutetium compounds. Lutetium chloride resulted in increases in prothrombin time (to more than 100 seconds by 100 minutes after treatment, compared to a maximum of 10 seconds for control animals) and coagulation time (to more than 60 minutes by 1 hour after treatment, compared with a maximum of about 10 minutes for control animals). No statistical analysis of these effects was provided in the report. Neither the citrate nor the edetate of lutetium resulted in comparable changes. The effect on clotting parameters was generally consistent among almost all of the rare-earth elements, especially the chloride compounds. Visual observation of pooled blood at incision sites provided additional evidence of the effect of rare-earth elements on clotting parameters, but the authors did not report the incidence or the specific treatment group(s) where this was observed. Gross and histopathological examinations revealed slight to moderate hyperemia of the lungs, only in animals treated with chlorides of the rare-earth elements. Nakamura et al. (1997) administered lutetium chloride intravenously to Wistar-KY rats. In each of three experiments, single doses of 10 or 20 mg Lu/kg were administered via the tail vein. In the first experiment, three rats/dose were sacrificed 2 hours after dosing and blood was collected to assess the partitioning of lutetium between serum and blood cell fractions. Lutetium was primarily found in the serum (85.8 and 83.9% for the low and high dose, respectively). In the second experiment, five rats/dose were sacrificed 1 day after dosing, and blood, liver, spleen, kidneys, and femurs were collected for analysis of lutetium and calcium content. Lutetium was primarily deposited in the liver (63.5 and 67.0%> for low and high doses, respectively) and bone (15.2 and 10.9%> for low and high doses, respectively). At the high dose, lutetium administration resulted in significantly (p<0.05) increased concentrations of calcium in the liver (3.7-fold), spleen (2.8-fold), lungs (1.3-fold) and kidneys (1.2-fold). In the final experiment, 3-5 rats were treated with 10 mg Lu/kg and sacrificed 1 or 3 days after dosing for evaluation of serum chemistry (aspartate aminotransferase, alanine aminotransferase, total cholesterol, phospholipids, triglycerides, total bile acids and bilirubin) and hepatic lipids (phospholipids, triglycerides and total cholesterol). Neither serum enzymes nor hepatic lipids were changed from control values in rats treated with lutetium chloride. The authors concluded that heavy rare-earth elements (including Lu) did not induce hepatotoxicity. No genotoxicity data for stable lutetium were located. 7 ------- 5-30-2007 DERIVATION OF PROVISIONAL SUBCHRONIC AND CHRONIC ORAL RfD VALUES FOR STABLE LUTETIUM Information on the toxicity of repeated oral exposure to lutetium is limited to a single subchronic dietary study in rats (Haley et al., 1964); this study was selected as the critical study for the p-RfD. No effects were observed on the parameters evaluated, so the highest of the three tested doses (504-590 mg/kg-day) was identified as a freestanding NOAEL. The only other information available on the oral toxicity of lutetium is an oral LD50 in male mice (4416 mg Lu/kg). The free-standing NOAEL reported by Haley et al. (1964) was used to derive a subchronic p-RfD of 0.5 mg/kg-day for lutetium. An uncertainty factor (UF) of 1000 was applied to the NOAEL to derive the subchronic p-RfD: Subchronic p-RfD = NOAEL / UF = 504 mg/kg-day / 1000 = 0.5 or 5E-1 mg/kg-day The UF of 1000 includes a factor of 10 for extrapolation from rats to humans, 10 for protection of sensitive individuals and 10 for deficiencies in the subchronic toxicity database, including lack of oral toxicity data in a second species, neurotoxicity data, given the overt neurotoxicology effects at high does, developmental and a multi-generation reproductive toxicity study. A chronic p-RfD was not derived for lutetium. There are no studies of chronic exposure to lutetium in any species. A study using radioactive lutetium reported substantial deposition of 177Lu to bone, and estimated a half-life of 2.5 years for elimination of heavier lanthanides (which would include lutetium) from the skeleton (Durbin et al., 1956). The potential for prolonged retention of lutetium in the body increases the uncertainty in extrapolating the toxicological effects observed in a subchronic study to effects after chronic exposure. Consequently, no chronic p-RfD was derived for lutetium. Confidence in the principal study is low. Although both sexes were tested in this study, a very small number of animals were used for each dose group (six per sex). The toxicological evaluation in this study was limited to body weight measures, selected hematological parameters, and histopathology of a subset of organs. Neither serum chemistry nor urinalysis endpoints were evaluated, nor were organ weight measurements made. An effect level (i.e., LOAEL) was not identified. Confidence in the subchronic database on lutetium is low. Apart from the critical study, the only other information on oral toxicity is an oral LD50 in mice. Oral absorption of lutetium is likely to be low based on limited data from other rare-earth elements (Durbin et al., 1956). There are no data to indicate the toxicological endpoint(s) or target organ(s) of subchronic or chronic oral exposure to lutetium. In addition, there is no information on potential neurotoxicity, apart from the observation of clinical signs of neurological effects (e.g., ataxia, sedation) at lethal doses in mice (Haley et al., 1964). No studies of the reproductive or developmental effects of stable lutetium are available. Studies using injection routes of exposure indicate that lutetium exposure may affect clotting parameters and that it can increase the calcium concentration in a number of organs. It is not clear whether these effects may occur 8 ------- 5-30-2007 after oral exposure, as they were not studied by Haley et al. (1964). Low confidence in the subchronic p-RfD follows. FEASIBILITY OF DERIVING PROVISIONAL SUBCHRONIC AND CHRONIC INHALATION RfCs FOR STABLE LUTETIUM No pertinent data regarding the toxicity of repeated inhalation exposure to stable lutetium were located in the available literature. Derivation of a p-RfC for stable lutetium is precluded by the lack of appropriate inhalation toxicity data. PROVISIONAL WEIGHT-OF-EVIDENCE CLASSIFICATION FOR STABLE LUTETIUM No carcinogenicity or genotoxicity data were located for stable lutetium. One subchronic oral toxicity study of lutetium chloride in rats was inadequate to assess carcinogenic potential. As the available data are insufficient to assess carcinogenic potential in animals or humans, they are consistent with the hazard descriptor, "inadequate information to assess carcinogenic potential," as specified by the U.S. EPA (2005) Cancer Guidelines. QUANTITATIVE ESTIMATES OF CARCINOGENIC RISK FOR STABLE LUTETIUM Derivation of quantitative estimates of cancer risk for lutetium is precluded by the lack of data demonstrating carcinogenicity associated with lutetium exposure. REFERENCES Abel, M. and R.B. Talbot. 1967. Gadolinium oxide inhalation by guinea pigs: A correlative functional and histopathologic study. J. Pharmacol. Exp. Therap. 157: 207-213. ATSDR (Agency for Toxic Substances and Disease Registry). 2006. Toxicological Profile Information Sheet. U.S. Department of Health and Human Services, Public Health Service. Atlanta, GA. Online, http://www.atsdr.cdc.gov/toxpro2.html. Accessed May 15, 2006. Ball, R.A. and G. VanGelder. 1966. Chronic toxicity of gadolinium oxide for mice following exposure by inhalation. Arch. Environ. Health. 13: 601-608. Beliles, R.P. 1994. The Lanthanides. In: Patty's Industrial Hygiene and Toxicology, Fourth edition, Volume 2, Part C. G.D. Clayton and F.E. Clayton, Ed. John Wiley and Sons, Inc., New York, NY. p. 2048-2065. 9 ------- 5-30-2007 Bruce, D.W, B.E. Hietbrink and K.P. DuBois. 1963. The acute mammalian toxicity of rare earth nitrates and oxides. Toxicol. Appl. Pharmacol. 5:750-759. Colombo, F., M. Zanoni and G. Vocaturo. 1983. Pneumoconiosi da terre rare. (Pneumoconiosis due to rare earth metals). Med. Lav. 74: 191-197. Deng, J.F., T. Sinks, L. Elliott et al. 1991. Characterisation of respiratory health and exposures at a sintered permanent magnet manufacturer. J. Ind. Med. 48: 609-615. Durbin P.W., M.H. Williams, M. Gee et al. 1956. Metabolism of the lanthanons in the rat. Proc. Soc. Exp. Biol. Med. 91:78-85. Graca J.G., F.C. Davison and J.B. Feavel. 1962. Comparative toxicity of stable rare earth compounds: II. Effect of citrate and edetate complexing on acute toxicity in mice and guinea pigs. Arch. Environ. Health. 5:437-450. Graca J.G., F.C. Davison and J.B. Feavel. 1964. Comparative toxicity of stable rare earth compounds: III. Acute toxicity of intravenous injections of chlorides and chelates in dogs. Arch. Environ. Health. 8:555-564. Haley, P.J. 1991. Pulmonary toxicity of stable and radioactive lanthanides. Health Physics. 61(6): 809-821. Haley, T.J., N. Komesu, M. Efros et al. 1964. Pharmacology and toxicology of lutetium chloride. J. Pharm. Sci. 53:1186-1188. Husain, M.H., J.A. Dick and Y.S. Kaplan. 1980. Rare earth pneumoconiosis. J. Soc. Occup. Med. 30: 15-19. IARC (International Agency for Research on Cancer). 2006. IARC Agents and Summary Evaluations. Online, http://www-cie.iarc.fr/. Accessed May 15, 2006. Kappenberger, L. and A.A. Buhlmann. 1975. Lungenveranderungen durch «seltene erden». (Lung lesions caused by "rare earths"). Schweiz. Med. Wochenschr. 105: 1799-1801. Mogilevskaya, O.Y. and N.T. Raikhlin. 1967. Rare Earth Metals. In: Izrael'son, Z.I. Ed. Toxicology of the Rare Earth Metals. Israel Program for Scientific Translations, Jerusalem, p. 132-141. Nakamura Y., Y.Tsumura, Y. Tonogai, et al. 1997. Differences in behavior among chlorides of seven rare earth elements administered intravenously to rats. Fundam. Appl. Toxicol. 37:106- 116. NTP (National Toxicology Program). 2006. Management Status Report. Online. http://ntp- server.niehs.nih.gov/. Accessed May 15,2006. 10 ------- 5-30-2007 Pharmacyclic. 1999. Pharmacyclic Announces Clinical Results with Lutetium Texaphyrin for Treatment of Age-Related Macular Degeneration. PRNewswire, July 28, 1999, pp 1048. Sabbioni, E., R. Pietra, P. Gaglione et al. 1982. Long-term occupational risk of rare-earth pneumoconiosis: A case report as investigated by neutron activation analysis. Sci. Total Environ. 26: 19-32. Schepers, G.W.H. 1955a. The biological action of rare earths. I. The experimental pulmonary histopathology produced by a blend having a relatively high oxide content. A.M. A. Arch. Ind. Health. 12: 301-305. Schepers, G.W.H. 1955b. The biological action of rare earths. II. The experimental pulmonary histopathology produced by a blend having a relatively high fluoride content. A.M. A. Arch. Ind. Health. 12:306-316. Schepers, G.W.H., A.B. Delahant and A.J. Redlin. 1955. An experimental study of the effects of rare earths on animal lungs. A.M. A. Arch. Ind. Health. 12: 297-300. Sulotto, F., C. Romano and A. Berra. 1986. Rare-earth pneumoconiosis: A new case. Am. J. Ind. Med. 9: 567-575. U.S. EPA. 1988. Recommendations for and Documentation of Biological Values for Use in Risk Assessment. Environmental Criteria and Assessment Office, Cincinnati, OH. NTIS PB179874. U.S. EPA. 1991. Chemical Assessments and Related Activities (CARA). Office of Health and Environmental Assessment, Washington, DC. U.S. EPA. 1994. Chemical Assessments and Related Activities (CARA). Office of Health and Environmental Assessment, Washington, DC. U.S. EPA. 1997. Health Effects Assessment Summary Tables. FY-1997 Update. Prepared by the Office of Research and Development, National Center for Environmental Assessment, Cincinnati OH for the Office of Emergency and Remedial Response, Washington, DC. EPA/540/R-97/036. NTIS PB97921199. U.S. EPA. 2004. 2004 Edition of the Drinking Water Standards and Health Advisories. Office of Water, Washington, DC. Winter, 2004. EPA 822-R-02-038. Online. http://www.epa.gov/waterscience/drinking/standards/dwstandards.pdf. Accessed May 15, 2006. U.S. EPA. 2005. Guidelines for carcinogen risk assessment. Risk Assessment Forum, Washington, DC; EPA/630/P-03/001F. Federal Register 70(66): 17765—17817. Available online at http://www.epa.gov/raf 11 ------- 5-30-2007 U.S. EPA. 2007. Integrated Risk Information System (IRIS). Office of Research and Development, National Center for Environmental Assessment, Washington, DC. Online. http://www.epa.gov/iris/. Accessed May 15,2006. Vocaturo, G., F. Colombo, M. Zanoni et al. 1983. Human exposure to heavy metals: Rare earth pneumoconiosis in occupational workers. Chest. 83:780-783. Vogt, P., M.A. Spycher and J.R. Ruttner. 1986. Pneumokoniose durch «seltene erden» (cer- pneumokoniose). [Pneumoconiosis caused by "rare earths" (cer-pneumoconiosis).] Schweiz. Med. Wochenschr. 116:1303-1308. Waring, P.M. and R.J. Watling. 1990. Rare earth deposits in a deceased movie projectionist: A new case of rare earth pneumoconiosis. Med. J. Aust. 153: 726-730. WHO (World Health Organization). 2006. Online Catalogs for the Environmental Criteria Series. Online, http://www.who.int/pcs/pubs/pub_ehc_alph.htm. Accessed May 15, 2006. 12 ------- |