https://ntp.niehs.nih.gov/go/tr507abs

Abstract for TR-507

Toxicology and Carcinogenesis Studies of Vanadium Pentoxide in F344/N Rats and B6C3F1 Mice (Inhalation Studies)

CASRN: 1314-62-1
Chemical Formula: V2O5
Molecular Weight: 181.88
Synonyms/Common Names: Divanadium pentoxide; vanadic anhydride; vanadium (5) oxide; vanadium oxide; vanadium pentaoxide
Report Date: December 2002

Full Report PDF

Abstract

Vanadium pentoxide, commercially the most important compound of vanadium, presents a potential occupational hazard during the cleaning of oil-fired boilers and furnaces, the handling of catalysts, and during the refining, processing, or burning of vanadium-rich mineral ores or fossil fuels. Vanadium pentoxide was nominated for study by the National Cancer Institute as a representative of the metals class study. Male and female F344/N rats and B6C3F1 mice were exposed to vanadium pentoxide (99% pure) by inhalation for 16 days, 14 weeks, or 2 years. Genetic toxicology studies were conducted in Salmonella typhimurium and mouse peripheral blood.

Sixteen-day study in rats

Groups of five male and five female rats were exposed to particulate aerosols of vanadium pentoxide at concentrations of 0, 2, 4, 8, 16, or 32 mg/m3 by inhalation, 6 hours per day, 5 days per week for 16 days. Three males in the 32 mg/m3 group died before the end of the study. Mean body weights of males and females exposed to 8 mg/m3 or greater were less than those of the chamber controls. Clinical findings included rapid respiration and hypoactivity in rats exposed to 16 or 32 mg/m3. Relative lung weights of 4 mg/m3 or greater males and 2 mg/m3 or greater females were significantly greater than those of the chamber controls. Lavage fluid analysis indicated an inflammatory response in the lung that was either directly mediated by vanadium pentoxide or was secondary to lung damage induced by vanadium pentoxide exposure.

Sixteen-day study in mice

Groups of five male and five female mice were exposed to particulate aerosols of vanadium pentoxide at concentrations of 0, 2, 4, 8, 16, or 32 mg/m3 by inhalation, 6 hours per day, 5 days per week for 16 days. All males exposed to 32 mg/m3 and one 8 mg/m3 male died or were killed moribund before the end of the study. Mean body weights of 16 mg/m3 males and 8 mg/m3 or greater females were significantly less than those of the chamber controls, and the 32 mg/m3 females lost weight during the study. Absolute and relative lung weights of 4 mg/m3 or greater males and all exposed groups of females and liver weights of 16 mg/m3 males were significantly greater than those of the chamber controls. The mediastinal lymph nodes were enlarged in 4, 8, and 16 mg/m3 males and females, and lymphoid hyperplasia was confirmed histologically. Lavage fluid analysis indicated an inflammatory response in the lung that was either directly mediated by vanadium pentoxide or was secondary to lung damage induced by vanadium pentoxide exposure.

 

Three-month study in rats

Groups of 10 male and 10 female rats were exposed to particulate aerosols of vanadium pentoxide at concentrations of 0, 1, 2, 4, 8, or 16 mg/m3 by inhalation, 6 hours per day, 5 days per week for 3 months. Seven males and three females exposed to 16 mg/m3 died during the study. Mean body weights were significantly less in males exposed to 4 mg/m3 or greater and in females exposed to 16 mg/m3 . Abnormal breathing, thinness, lethargy, abnormal posture, and ruffled fur were observed in rats exposed to 16 mg/m3.

Hematology results indicated that exposure of rats to vanadium pentoxide induced a microcytic erythrocytosis in males and females. Absolute and relative lung weights were significantly greater for 4 mg/m3 or greater males and females than for the chamber controls as were the relative lung weights of 2 mg/m3 males. The estrous cycle of females exposed to 8 mg/m3 was significantly longer than that of the chamber control group, and the number of cycling females in the 16 mg/m3 group was reduced. The incidences of several nonneoplastic lesions of the lung and nose were significantly increased in males and females exposed to 2 mg/m3 or greater. Data from pulmonary function analyses indicated that a restrictive lung disease was present in male and female rats exposed to 4 mg/m3 or greater, while an obstructive lung disease was present only in the 16 mg/m3 groups.

Three-month study in mice

Groups of 10 male and 10 female mice were exposed to particulate aerosols of vanadium pentoxide at concentrations of 0, 1, 2, 4, 8, or 16 mg/m3 by inhalation, 6 hours per day, 5 days per week for 3 months. One male exposed to 16 mg/m3 died before the end of the study. Mean body weights of 8 and 16 mg/m3 males and 4 mg/m3 or greater females were significantly less than those of the chamber controls. Absolute and relative lung weights of males and females exposed to 4 mg/m3 or greater were significantly greater than those of the chamber controls. The epididymal spermatozoal motility of males exposed to 8 or 16 mg/m3 was significantly decreased. Some mice exposed to 2 or 4 mg/m3 had inflammation of the lung, and all mice exposed to 8 or 16 mg/m3 had inflammation and epithelial hyperplasia of the lung.

Sixteen-day special study in rats

Groups of 60 female rats were exposed to particulate aerosols of vanadium pentoxide at concentrations of 0, 1, or 2 mg/m3 and groups of 40 female rats were exposed to 4 mg/m3 by inhalation, 6 hours per day, 5 days per week for 16 days. Alveolar and bronchiolar epithelial hyperplasia was observed in most rats exposed to 2 or 4 mg/m3 on days 6 and 13. Histiocytic infiltration and inflammation occurred in a time- and concentration-related manner. Cell turnover rates were increased in the terminal bronchioles on days 6 and 13 and in the alveoli in the 4 mg/m3 group on day 6 and in all exposed groups on day 13. Assessment of lung vanadium concentrations suggested deposition and clearance exhibited linear kinetics over the exposure range studied. Lung clearance half-times ranged from 4.42 to 4.96 days.

Sixteen-day special study in mice

Groups of 60 female mice were exposed to particulate aerosols of vanadium pentoxide at concentrations of 0, 2, or 4 mg/m3 and groups of 40 female mice were exposed to 8 mg/m3 by inhalation, 6 hours per day, 5 days per week for 16 days. Alveolar and bronchiolar epithelial hyperplasia occurred with similar incidences and severities among the exposed groups on days 6 and 13, and time- and concentration-related increases in the incidences of interstitial inflammation and histiocytic infiltration also occurred in these groups. Cell turnover rates were increased in the terminal bronchioles on day 6 and remained greater than those of the chamber controls on day 13. In the alveoli, cell turnover rates were increased in an exposure concentration-related manner on day 13; cell turnover rates were increased only in the 8 mg/m3 group on day 6. Assessment of lung vanadium concentrations suggested deposition and clearance exhibited linear kinetics over the exposure range studied. Lung clearance half-times ranged from 2.40 to 2.55 days.

Two-year study in rats

Groups of 50 male and 50 female rats were exposed to particulate aerosols of vanadium pentoxide at concentrations of 0, 0.5, 1, or 2 mg/m3 by inhalation, 6 hours per day, 5 days per week for 104 weeks. Survival and body weights of males and females were generally similar to those of the chamber controls. Mean body weights of females exposed to 2 mg/m3 were less than those of the chamber controls throughout the study.

Alveolar/bronchiolar neoplasms were present in exposed groups of male rats, and the incidences often exceeded the historical control ranges. Alveolar/bronchiolar adenomas were present in 0.5 and 1 mg/m3 females; one 2 mg/m3 female also had an alveolar/bronchiolar carcinoma. The incidence of alveolar/bronchiolar adenoma in the 0.5 mg/m3 group was at the upper end of the historical control ranges. Nonneoplastic lesions related to vanadium pentoxide exposure occurred in the respiratory system (lung, larynx, and nose) of male and female rats, and the severities of these lesions generally increased with increasing exposure concentration.

Two-year study in mice

Groups of 50 male and 50 female mice were exposed to particulate aerosols of vanadium pentoxide at concentrations of 0, 1, 2, or 4 mg/m3 by inhalation, 6 hours per day, 5 days per week for 104 weeks. Survival of 4 mg/m3 males was significantly less than that of the chamber controls. Mean body weights of 4 mg/m3 males and all exposed groups of females were generally less than those of the chamber controls throughout the study, and those of males exposed to 2 mg/m3 were less from week 85 to the end of the study. Many mice exposed to vanadium pentoxide were thin, and abnormal breathing was observed in some mice, particularly those exposed to 2 or 4 mg/m3.

The incidences of alveolar/bronchiolar neoplasms were significantly increased in all groups of exposed males and females. Nonneoplastic lesions related to vanadium pentoxide exposure occurred in the respiratory system (lung, larynx, and nose) of male and female mice, and the severities of these lesions generally increased with increasing exposure concentration. Bronchial lymph node hyperplasia was present in many exposed females.

Molecular oncology studies

K-ras codon 12 mutation and loss of heterozygosity on chromosome 6 were detected in vanadium pentoxide-induced alveolar/bronchiolar carcinomas from mice.

Genetic toxicology

Vanadium pentoxide was not mutagenic in Salmonella typhimurium strain TA97, TA98, TA100, TA102, or TA1535, with or without induced rat or hamster liver S9 enzymes. Vanadium pentoxide, administered for 3 months by inhalation to male and female mice, did not increase the frequency of micronucleated normochromatic erythrocytes in peripheral blood.

Conclusions

Under the conditions of this 2-year inhalation study, there was some evidence of carcinogenic activity of vanadium pentoxide in male F344/N rats and equivocal evidence of carcinogenic activity of vanadium pentoxide in female F344/Nrats based on the occurrence of alveolar/bronchiolar neoplasms. There was clear evidence of carcinogenic activity of vanadium pentoxide in male and female B6C3F1 mice based on increased incidences of alveolar/bronchiolar neoplasms.

Exposure to vanadium pentoxide caused a spectrum of nonneoplastic lesions in the respiratory tract (nose, larynx, and lung) including alveolar and bronchiolar epithelial hyperplasia, inflammation, fibrosis, and alveolar histiocytosis of the lung in male and female rats and mice and an unusual squamous metaplasia of the lung in male and female rats. Hyperplasia of the bronchial lymph node occurred in female mice.

Studies

Summary of Two-year Carcinogenesis and Genetic Toxicology Studies of Vanadium Pentoxide

 

Male
F344/N Rats
Female
F344/N Rats
Male
B6C3F1 Mice
Female
B6C3F1 Mice
Concentration in air

Chamber control, 0.5, 1, or 2 mg/m3

Chamber control, 0.5, 1, or 2 mg/m3

Chamber control, 1, 2 or 4 mg/m3

Chamber control, 1, 2 or 4 mg/m3

Body weights

Exposed groups similar to the chamber control group

2 mg/m3 group less than the chamber control group

2 and 4 mg/m3 group less than the chamber control group

Exposed groups less than the chamber control group

Survival rates

20/50, 29/50, 26/50, 27/50

33/50, 24/50, 29/50, 30/50

39/50, 33/50, 36/50, 27/50

38/50, 32/50, 30/50, 32/50

Nonneoplastic effects

Lung: alveolar epithelium, hyperplasia (7/50, 24/49, 34/48, 49/50); bronchiole, epithelium hyperplasia (3/50, 17/49, 31/48, 49/50); alveolar epithelium, metaplasia, squamous (1/50, 0/49, 0/48, 21/50); bronchiole, metaplasia, squamous (0/50, 0/49, 0/48, 7/50); inflammation, chronic active (5/50, 8/49, 24/48, 42/50); interstitial, fibrosis (7/50, 7/49, 16/48, 38/50); alveolus, infiltration cellular, histiocyte (22/50, 40/49, 45/48, 50/50)

Larynx: inflammation, chronic (3/49, 20/50, 17/50, 28/49); respiratory epithelium, epiglottis degeneration (0/49, 22/50, 23/50, 33/49); respiratory epithelium, epiglottis, hyperplasia (0/49, 18/50, 34/50, 32/49); respiratory epithelium, epiglottis, metaplasia, squamous (0/49, 9/50, 16/50, 19/49)

Nose: goblet cell, respiratory epithelium, hyperplasia (4/49, 15/50, 12/49, 17/48)

Lung: alveolar epithelium, hyperplasia (4/49, 8/49, 21/50, 50/50); bronchiole, epithelium hyperplasia (6/49, 5/49, 14/50, 48/50); alveolar epithelium, metaplasia, squamous (0/49, 0/49, 0/50, 6/50); inflammation, chronic active (10/49, 10/49, 14/50, 40/50); interstitial, fibrosis (19/49, 7/49, 12/50, 32/50); alveolus, infiltration cellular, histiocyte (26/49, 35/49, 44/50, 50/50)

Larynx: inflammation, chronic (8/50, 26/49, 27/49, 37/50); respiratory epithelium, epiglottis degeneration (2/50, 33/49, 26/49, 40/50); respiratory epithelium, epiglottis, hyperplasia (0/50, 25/49, 26/49, 33/50); respiratory epithelium, epiglottis, metaplasia, squamous (2/50, 7/49, 7/49, 16/50)

Nose: goblet cell, respiratory epithelium, hyperplasia (13/50, 18/50, 16/50, 30/50)

Lung: alveolar epithelium, hyperplasia (3/50, 41/50, 49/50, 50/50); bronchiole, epithelium, hyperplasia (0/50, 15/50, 37/50, 46/50); inflammation chronic (6/50, 42/50, 45/50, 47/50); alveolus, infiltration cellular, histiocyte (10/50, 36/50, 45/50, 49/50); interstitial fibrosis (1/50, 6/50, 9/50, 12/50)

Larynx: respiratory epithelium, epiglottis, metaplasia, squamous (2/49, 45/50, 41/48, 41/50)

Nose: inflammation suppurative (16/50, 11/50, 32/50, 23/50); olfactory epithelium, degeneration, hyaline (1/50, 7/50, 23/50, 30/50); respiratory epithelium, degeneration, hyaline (8/50, 22/50, 38/50, 41/50); respiratory epithelium, metaplasia, squamous (0/50, 6/50, 6/50, 2/50)

Lung: alveolar epithelium, hyperplasia (0/50, 31/50, 38/50, 50/50); bronchiole, epithelium, hyperplasia (0/50, 12/50, 34/50, 48/50); inflammation chronic (4/50, 37/50, 39/50, 49/50); alveolus, infiltration cellular, histiocyte (0/50, 34/50, 35/50, 45/50); interstitial fibrosis (0/50, 1/50, 4/50, 8/50)

Larynx: respiratory epithelium, epiglottis, metaplasia, squamous (0/50, 39/50, 45/49, 44/50)

Nose: inflammation suppurative (19/50, 14/50, 32/50, 30/50); olfactory epithelium, atrophy (2/50, 8/50, 5/50, 14/50); olfactory epithelium, degeneration, hyaline (11/50, 23/50, 34/50, 48/50); respiratory epithelium, degeneration, hyaline (35/50, 39/50, 46/50, 50/50); respiratory epithelium, metaplasia, squamous (0/50, 3/50, 7/50, 8/50); respiratory epithelium, necrosis (0/50, 0/50, 1/50, 7/50)

Bronchial lymph node: hyperplasia (3/39, 13/40, 14/45, 20/41)

 

Neoplastic effects

Lung: alveolar/bronchiolar adenoma (4/50, 8/49, 5/48, 6/50); alveolar/bronchiolar carcinoma (0/50, 3/49, 1/48, 3/50); alveolar/bronchiolar adenoma or carcinoma (4/50, 10/49, 6/48, 9/50)

None

Lung: alveolar/bronchiolar adenoma (13/50, 16/50, 26/50, 15/50); alveolar/bronchiolar carcinoma (12/50, 29/50, 30/50, 35/50); alveolar/bronchiolar adenoma or carcinoma (22/50, 42/50, 43/50, 43/50)

Lung: alveolar/bronchiolar adenoma (1/50, 17/50, 23/50, 19/50); alveolar/bronchiolar carcinoma (0/50, 23/50, 18/50, 22/50); alveolar/bronchiolar adenoma or carcinoma (1/50, 32/50, 35/50, 32/50)

Equivocal findings

None

Lung: alveolar/ bronchiolar adenoma (0/49, 3/49, 1/50, 0/50); alveolar/bronchiolar adenoma or carcinoma (0/49, 3/49, 1/50, 1/50)

None

None

Levels of evidence of carcinogenic activity

Some evidence

Equivocal evidence

Clear evidence

Clear evidence

Genetic Toxicology
Assay Results
Salmonella typhimurium gene mutations: Negative in strains TA97, TA98, TA100, TA102 and TA1535 with and without S9
Micronucleated erythrocytes
Mouse peripheral blood in vivo:
Negative