Registration Dossier

Data platform availability banner - registered substances factsheets

Please be aware that this old REACH registration data factsheet is no longer maintained; it remains frozen as of 19th May 2023.

The new ECHA CHEM database has been released by ECHA, and it now contains all REACH registration data. There are more details on the transition of ECHA's published data to ECHA CHEM here.

Diss Factsheets

Administrative data

Endpoint:
immunotoxicity
Remarks:
other: review of various in vivo studies
Type of information:
migrated information: read-across based on grouping of substances (category approach)
Adequacy of study:
key study
Reliability:
2 (reliable with restrictions)
Rationale for reliability incl. deficiencies:
other: A Concise International Chemical Assessment Document (CICAD) on cyclic acid anhydrides was written by expert groups in occupational health and safety, with peer review and final board approval.

Data source

Reference
Reference Type:
review article or handbook
Title:
Cyclic Acid Anhydrides: Human Health Aspects.
Author:
World Health Organization
Year:
2009
Bibliographic source:
International Chemical Assessment Document 75: 1-63. http://www.inchem.org/documents/cicads/cicads/cicad75.pdf

Materials and methods

Test guideline
Qualifier:
no guideline followed
Principles of method if other than guideline:
The review article presents investigations of in vivo studies of immune response to anhydrides
GLP compliance:
no
Limit test:
no

Test material

Constituent 1
Chemical structure
Reference substance name:
Benzene-1,2,4-tricarboxylic acid 1,2-anhydride
EC Number:
209-008-0
EC Name:
Benzene-1,2,4-tricarboxylic acid 1,2-anhydride
Cas Number:
552-30-7
Molecular formula:
C9H4O5
IUPAC Name:
1,3-dioxo-1,3-dihydro-2-benzofuran-5-carboxylic acid
Constituent 2
Reference substance name:
trimellitic anhydride
IUPAC Name:
trimellitic anhydride
Details on test material:
no data

Test animals

Species:
other: various
Details on test animals or test system and environmental conditions:
review of in vivo studies

Administration / exposure

Route of administration:
other: various
Details on exposure:
review of in vivo studies
Analytical verification of doses or concentrations:
not specified

Results and discussion

Results of examinations

Details on results:
Numerous studies indicate that dermal and respiratory sensitisation reactions after trimellitic anhydride exposure are mediated through IgE and IgG antibody formation.

Effect levels

Dose descriptor:
other:
Effect level:
other: various experimental conditions
Based on:
test mat.
Sex:
male/female
Basis for effect level:
other: The results of numerous studies indicate that TMA is associated with IgE and IgG mediated immune responses after exposure to anhydrides.

Any other information on results incl. tables

Antibody-mediated sensitization studies:

 

The studies described in this section demonstrate that antibody responses have been induced by cyclic acid anhydrides via bronchial, subcutaneous, intradermal, and parenteral routes of exposure. Development of an allergic respiratory disease is dependent on the production of specificantibodies.

 

Zeiss et al. (1987) conducted an inhalation experi- ment by exposing rats to trimellitic anhydride dust at concentrations of 0, 10, 30, 100, or 300 µg/m3for 6 h/day for 5 or 10 days. Exposure levels of 30–300 µg/m3for 10 days caused haemorrhagic lung foci. Anti-trimellitic anhydride–rat serum albumin antibody binding was correlated with exposure concentration, the presence of haemorrhagic lung foci, and lung weight.By 12 days post-exposure, the lung lesions healed, although a repeated exposure caused a return of lesions (Zeiss et al., 1987). Histological evaluation of the lung lesions indicated extensive cellular infiltration of primarily macrophages, alveolar haemorrhage, andpneumonitis. These effects presented in a dose-dependent manner. The lungs were the only affected organs (Leach et al., 1987).

 

Chandler et al. (1987) exposed rats by inhalation to trimellitic anhydride powder at a concentrationof

100 µg/m3for 6 h/day, 5 days/week, for 2 weeks. At autopsy, the surface of the lungs had haemorrhagicfoci.

Higher total antibody concentrations were observed in the bronchoalveolar lavage fluid than in serum. Anti- trimellitic anhydride–rat serum albumin IgG, IgA, and IgM were detected (Chandler et al., 1987). Antibody levels in bronchoalveolar lavage and serum were highly correlated with lung injury (Zeiss et al.,1988).

 

In two separate studies, Zeiss et al. (1989) exposed rats by inhalation to trimellitic anhydride powder at concentrations of 330 or 500 µg/m3 on days 1, 5, and 10 for 6 h/day and challenged the rats with trimellitic anhydride at 300 or 540 µg/m3on day 22 or day 29, respectively. In the 500 µg/m3exposure group, anti- trimellitic anhydride–rat serum albumin IgM and IgA began increasing on day 5 and peaked on day 20.IgG

antibodies began increasing on day 7 and also peaked on day 20. Rats of the low exposure group (330 µg/m3) that were not rechallenged had fewer lung foci than the rechallenged rats. The rechallenged rats also demonstrated a strong correlation between antibody measures and lung injury. A subgroup of rats was exposedto

500 µg/m3on days 1 and 5 and challenged on day29 with the same concentration. A good correlation between antibody response and lung injury was observed.

 

Zhang et al. (2006) exposed Brown Norway rats to trimellitic anhydride aerosol at concentrations of 0.04, 0.4, 4, or 40 mg/m3for 10 min, once per week, for over 10 weeks. The rats were then challenged withtrimellitic anhydride aerosol at 40 mg/m3. Rats sensitized in the 40 mg/m3group developed specific IgE and both early- phase and late-phase airway responses. Rats inthe 4 mg/m3group exhibited a lower but stable specific IgE response; early-phase and late-phase airway responses were observed only after the 40 mg/m3challenge and were greater than those observed in the 40mg/m3 sensitization group. Histopathological changes were exposure dependent and included eosinophilic granulomatous interstitial pneumonia, perivascular eosinophil infiltrates, bronchial-associated lymphoid tissue hyperplasia, and peribronchiolar plasma cellinfiltrates.

 

Dykewicz et al. (1988) sensitized two rhesus monkeys intrabronchially with serum from a human worker who had trimellitic anhydride asthma and high titres of anti-trimellitic anhydride–human serum albumin IgE, IgG, and IgA. The monkeys were challengedwith trimellitic anhydride–human serum albumin aerosol and developed bronchospasm. After 1 week, the challenge was negative. Passive cutaneous anaphylaxis (using the Prausnitz-Küstner test) waspositive.

 

Hayes et al. (1992a) developed a guinea-pig model for trimellitic anhydride–induced airway hypersensi- tivity. Guinea-pigs were sensitized intradermallywith 0.1 ml of 0.3% trimellitic anhydride in corn oil. Specific serum IgG1antibody levels were increased in all sensi- tized animals. IgE antibodies were detected in six out of eight sensitized animals. On days 21–28, guinea-pigs were challenged with a tracheal dose of 50 µl of 1% trimellitic anhydride–guinea-pig serum albumin, which caused increased lung resistance in sensitized animals compared with non-sensitized animals. Evans blue testing revealed airway microvascular leakage in sensi- tized guinea-pigs. Challenge by nose inhalation of trimellitic anhydride at 12 000 µg/m3for 30 min resulted in a significant increase in bronchial reactivity at 8 h post-exposure, which was accompanied by an eosinophilic inflammatoryexudate.

 

Arakawa et al. (1993b) sensitized guinea-pigs by two intradermal injections of 0.1 ml of 0.3% trimellitic anhydride in corn oil and evaluated the time course of immune and airway responses. Animals were challenged with 50 µl of 0.5% trimellitic anhydride–guinea-pig serum albumin at 1, 2, 3, 5, and 8 weeks post-sensitization. The challenge induced significant increases in lung resistance, which reached a maximum at 2.5 min in the 1-week group and between 5 and 6 min in the other groups. Significant extravasation was observed, which increased up to 8 weeks. Specific IgG1antibodies were detected in all guinea-pigs of the 3-, 5-, and 8-week groups, which correlated with extravasation but not with increased lungresistance.

 

Zhang et al. (1998b) found specific IgE and IgG antibodies induced in intradermal studies of phthalic anhydride, trimellitic anhydride, maleic anhydride, hexahydrophthalic anhydride, methyl hexahydrophthalic anhydride, and methyl tetrahydrophthalicanhydride.

 

Cui et al. (1997) sensitized Brown Norway rats intradermally with trimellitic anhydride and then challenged the rats either once or 7 times withtrimellitic anhydride–rat serum albumin. Anti-trimellitic anhydride IgE and IgG were observed at high levels in all sensitized rats compared with controls. Repeated challenges with allergen, but not single challenges, caused significant bronchial hyperreactivity in sensitized rats. For example, repeated low-dose challenges produced greater hyperreactivity than a single 10-fold higher dose. Sensi- tized and single-challenged rats exhibited bronchial eosinophilia, although the non-sensitized non-challenged and sensitized rechallenged rats didnot.

 

Arts et al. (1998) challenged intradermally sensitized Brown Norway rats with trimellitic anhydride by inhalation, which induced immediate bronchoconstriction. Sensitized rats also exhibited eosinophilic aggre- gates, goblet cell hyperplasia and hypertrophy in the lungs, and the induction of haemorrhages. Non-sensitized rats exhibited less marked eosinophilic infiltration of the lungs after challengetests.

 

Arts et al. (2004) investigated airway responses of sensitized Brown Norway and Wistar rats to trimellitic anhydride. Rats were sensitized by dermal applications of 50% weight by volume (w/v) and then 25% w/v trimellitic anhydride. All rats were challenged 3 weeks after the first sensitization to a range of trimellitic anhydride concentrations (0.2–61 mg/m3for Brown Norway rats; 15–250 mg/m3for Wistar rats). Sensitized Brown Norway rats displayed elevated total IgE levels; inhalation challenge with2 mg/m3caused laryngeal inflammation with squamous epithelial metaplasia and pulmonary haemorrhages. Decreased breathing frequency and altered breathing patterns were concentration related. Inhalation challengeswith12 mg/m3caused increased lung weight. Nonspecific airway responsiveness was increased at 46and 61 mg/m3. The non-sensitized Brown Norwayrats displayed laryngeal squamous metaplasia (at higher challenge concentrations), decreased breathing frequency, and a breathing pattern characteristic of irritation. Sensitized Wistar rats exhibited airway inflammation and pulmonary haemorrhages upon challenge, but no functional changes, even at the highest concentrations that cause irritation. The authors concluded that the lowest NOEL was 0.2mg/m3.

 

Pauluhn (2003) performed a dose–response analysis and time course evaluation for intradermally sensitized Brown Norway rats challenged by inhalation. Sensitization was performed using 1, 5, or 25% trimellitic anhy- dride in acetone/olive oil that was applied 2 times, 1 week apart. Inhalation challenges were performed at 25– 30 mg trimellitic anhydride/m3for 30 min on days 17, 24, 41, 47, 55, and 66. Breathing patterns were altered only in the rats sensitized with 5% or 25% trimellitic anhydride. These rats also demonstrated an increased responsiveness to methacholine aerosol challengethe day following trimellitic anhydride challenge (onlyon day 17). The concentration of 5% trimellitic anhydride was determined to be the minimal sensitizing concentration. A time-related increase in airway responsiveness was observed. After the last challenge (day 66), the respiratory response and lung weights were similar to those observed in the repetitively rechallenged control group (non-sensitizedintradermally).

 

Dearman & Kimber (1991) developed a mouse model to differentiate chemicals for different types of allergenicity. Mice were topically sensitized by application of the test chemical in 4:1 acetone:olive oil to a shaved flank under an occluded patch for 48 h. Ear thickness was measured after 5 days, and the dorsum of both ears was treated with 25 µl of the test chemicals, trimellitic anhydride and 2,4-dinitrochlorobenzene. 2,4- Dinitrochlorobenzene is a potent contact allergen that lacks respiratory sensitization properties. Trimellitic anhydride and 2,4-dinitrochlorobenzene induced comparable levels of contact sensitization and antihapten IgG. However, only trimellitic anhydride induced IgE production. In addition, trimellitic anhydride induced IgG2brather than IgG2a, whereas the opposite was observed for 2,4-dinitrochlorobenzene. This may have been due to differences in T lymphocyte responses to the chemicals (Th1versus Th2). Similar responses have been observed with phthalic anhydride, maleic anhydride, hexahydrophthalic anhydride, and methyl tetrahydro- phthalic anhydride (Dearman & Kimber, 1992; Dearman et al., 2000). Arts et al. (1997) conducted similar experiments in Brown Norway rats usingtrimellitic anhydride, dinitrochlorobenzene, formaldehyde, andmethyl salicylate. A significant increase in serum IgE was observed after trimellitic anhydride exposure, but not after exposure to the otherchemicals.

 

Animal studies using blocking agents have demonstrated that histamine and thromboxane A2are primarily responsible for the early and late bronchoconstriction responses to trimellitic anhydride (Hayes et al., 1992b, 1995; Arakawa et al., 1993a, 1994b). Leukotrienes and histamine were found to mediate airwayexudation.

Pretreatment of sensitized guinea-pigs with budesonide, an anti-inflammatory corticosteroid, significantly inhibited the increase in airway responsiveness, but not the eosinophilic inflammation caused by exposure to trimellitic anhydride dust (Hayes et al., 1993). Rats pretreated with cyclophosphamide, an immunosuppressant, did not develop lung lesions or antibody responses after exposure to trimellitic anhydride at95 µg/m3, 6 h/day, 5 days/week, for 2 weeks (Leach etal., 1988). This study demonstrated that elimination of Tand B lymphocyte function could prevent trimellitic anhydride–induced lesions. Pretreatment of guinea-pigs with cyclosporin A caused inhibition of trimellitic anhydride–induced immunization processes; beta-methasone and azelastine did not cause inhibition (Arakawa et al., 1994a). However, a study by Pullerits et al. (1997) in Brown Norway rats demonstrated that both betamethasone and cyclosporin A administered during the sensitization period inhibited development of trimellitic anhydride–specific IgE andIgG.

 

Yan et al. (1995) demonstrated that sensitized guinea-pigs underwent activation of inducible nitric oxide synthase in bronchial tissue after challenge with trimellitic anhydride–guinea-pig serumalbumin.

 

Fraser et al. (1995) pretreated guinea-pigs with cobra venom, which reduced complement component C3in bronchoalveolar lavage fluid after challenge with trimellitic anhydride–guinea-pig serum albumin. Cobra venom pretreatment did not affect immediate bronchoconstriction and microvascular leakage. However, cobra venom pretreatment significantly reducedtrimellitic anhydride–induced increases in mononuclear cells, total white blood cells and red blood cells, and erythrocyte peroxidase activity. These results indicated that inhibition of complement activation by cobra venom prevented inflammatory cell infiltration in trimellitic anhydride–inducedasthma.

 

Larsen et al. (2001) also investigated the role of the complement system in trimellitic anhydride–induced allergic response in guinea-pig lung. Guinea-pigs were sensitized by intradermal injection of trimellitic anhydride. The complement activation product C3awas detected in bronchoalveolar lavage of both sensitized and non-sensitized guinea-pigs after intratracheal challenge with trimellitic anhydride–guinea-pig serum albumin. In sensitized animals, this challenge caused significant increases in eosinophils, neutrophils, and macrophages in lung and increases in red blood cells and protein in airspace. In a follow-up study, Larsen & Regal (2002) used 1 mg trimellitic anhydride dust for the challenge rather than trimellitic anhydride conjugated to guinea-pig serum albumin. The dust challenge was delivered by intratracheal insufflation. The non- sensitized guinea-pigs displayed significant increases in pulmonary resistance and decreases in dynamic lung compliance and blood pressure after challenge. The sensitized animals displayed significantly greater effects compared with the non-sensitized animals. In both sensitized and non-sensitized guinea-pigs, the dust challenge caused increased eosinophils in the lung tissues. This study demonstrates that trimellitic anhydride dust causes significant airway obstruction and eosinophilia in non-sensitized animals, and even greater effects in sensitizedanimals.

 

Valstar et al. (2006a) investigated the role of alveolar macrophages in asthma-like symptoms caused by trimellitic anhydride. Female Brown Norway rats were sensitized by dermal application of trimellitic anhydride on days 0 and 7. The day prior (day 20) to inhalation challenge with trimellitic anhydride (day 21), the rats were treated intratracheally with either empty liposomes (sham control) or liposomes containing clodronate (dichloromethylene diphosphonate) to deplete the lungs of alveolar macrophages. The sensitized rats exhibited decreased lung function parameters during and within 1 h after challenge compared with non-sensitized rats. Depletion of alveolar macrophages alleviated the trimellitic anhydride–induced decrease in lung function parameters and caused a quicker recovery compared with the sham controls; however, trimellitic anhydride– induced tissue damage and inflammation 24 h after challenge were augmented. This study concluded that alveolar macrophages have a dual role, since they potentiate the immediate decrease in lung function but suppress the inflammatory reaction 24 h later. Valstar et al. (2006b) conducted the same study but performed the inhalation challenge with trimelliticanhydride–bovine in the sensitized rats compared with the non-sensitized rats, irrespective of alveolar macrophage depletion. In addition, the challenge induced airway inflammation and tissue damage only when alveolar macrophages were depleted, irrespective of sensitization. These data indicate that alveolar macrophages inhibit nonspecific damage and inflammatory cell influx into the lungs caused by trimellitic anhydride–bovine serum albumin challenge.

 

Regal et al. (2001) evaluated eosinophil infiltration into lungs of BALB/c mice sensitized intradermallywith 0.1 ml of either 3% trimellitic anhydride or 0.3% ovalbumin (positive control). These mice were challenged 3 weeks later with 30 or 400 µg trimellitic anhydride– mouse serum albumin or 30 µg ovalbumin by intra- tracheal instillation. The numbers of eosinophils andneutrophils in bronchoalveolar lavage fluid were determined by eosinophil peroxidase andmyeloperoxidase activity, respectively. In the trimellitic anhydride– sensitized mice, the trimellitic anhydride–mouse serum albumin challenge resulted in a significant increase in eosinophil peroxidase activity. A small, but significant, increase was also observed in the non-sensitized mice. Total IgE in plasma and bronchoalveolar lavage fluid was significantly higher in trimellitic anhydride– sensitized mice than in the non-sensitized mice. The magnitudes of these responses were similar to those elicited by ovalbumin sensitization andchallenge.

Applicant's summary and conclusion

Conclusions:
The WHO expert group evaluated many experiments with sensitized animals after exposure to anhydrides, which demonstrated the formation of anhydride-specific IgE and IgG antibodies. Additionally, cytokines, complement and other immunological processes have been shown to be involved in anhydride-mediated immune reactions. This study is informative for evaluation of the toxicity of members of the cyclic acid anhydride category.