NCBI Bookshelf. A service of the National Library of Medicine, National Institutes of Health.
IARC Working Group on the Evaluation of Carcinogenic Risks to Humans. Pharmaceuticals. Lyon (FR): International Agency for Research on Cancer; 2012. (IARC Monographs on the Evaluation of Carcinogenic Risks to Humans, No. 100A.)
Tamoxifen was considered by a previous IARC Working Group in 1996 (IARC, 1996). Since that time, new data have become available, these have been incorporated into the Monograph, and taken into consideration in the present evaluation.
1. Exposure Data
1.1. Identification of the agent
1.1.1. Tamoxifen
- Chem. Abstr. Serv. Reg. No.: 10540-29-1
- Chem. Abstr. Name: (Z)-2-[4-(1,2-Diphenyl-1-butenyl)phenoxy]-N,N-dimethylethanamine
- IUPAC Systematic Name: 2-[4-[(Z)-1,2-Diphenylbut-1-enyl]phenoxy]-N,N-dimethylethanamine
- Synonyms: 1-p-β-Dimethylaminoethoxyphenyl-trans-1,2-diphenylbut-1-ene; (Z)-2-[4-(1,2-diphenylbut-1-enyl)phenoxy]ethyldimethylamine
- Description: Crystalline solid (O’Neil, 2006)
(a) Structural and molecular formulae, and relative molecular mass

C26H29NO
Relative molecular mass: 371.52
1.1.2. Tamoxifen citrate
- Chem. Abstr. Serv. Reg. No.: 54965-24-1
- Chem. Abstr. Name: (Z)-2-[4-(1,2-Diphenyl-1-butenyl)phenoxy]-N,N-dimethylethanamine, 2-hydroxy-1,2,3-propanetricarboxylate (1:1)
- IUPAC Systematic Name: 2-[4-[(Z)-1,2-Diphenylbut-1-enyl]phenoxy]-N,N-dimethylethylamine; 2-hydroxypropane-1,2,3-tricarboxylic acid
- Synonyms: Kessar; Nolvadex; Soltamox; tamoxifen citrate; Z-tamoxifen citrate
- Description: Fine, white, odourless crystalline powder (O’Neil, 2006)
(a) Structural and molecular formulae, and relative molecular mass

C26H29NO.C6H8O7
Relative molecular mass: 563.64
1.2. Use of the agent
Information for Section 1.2 is taken from IARC (1996), AstraZeneca Pharmaceuticals LP (2005), Royal Pharmaceutical Society of Great Britain (2007), and Thomson Healthcare (2007).
1.2.1. Indications
Tamoxifen has been available since the early 1970s for the first-line treatment of metastatic breast cancer in postmenopausal women. Tamoxifen has also been used as adjuvant therapy for treatment of postmenopausal, node-positive women with positive estrogen-receptor or progesterone-receptor levels and, since the early 1990s, for the treatment of postmenopausal node-negative women with positive estrogen-receptor or progesterone-receptor levels. In the late 1980s and early 1990s, it was also widely used in treating postmenopausal receptor-negative women (IARC, 1996; Wolff & Abeloff, 2002; Albain, 2004).
In women with ductal carcinoma in situ following breast surgery and radiation therapy, tamoxifen is used to reduce the risk of invasive breast cancer.
Tamoxifen has been considered as a chemopreventive agent to reduce the incidence of breast cancer in women at high risk of breast cancer.
Tamoxifen has been used as the first-line therapy for hormone-responsive male breast cancer, and is also used as adjuvant therapy for estrogen receptor- or progesterone receptor-positive male breast cancer.
Tamoxifen has also been used for anovulatory infertility.
1.2.2. Dosage
Tamoxifen is available as 10 mg and 20 mg tablets (each tablet contains 15.2 mg and 30.4 mg, respectively, of tamoxifen citrate and as an oral solution (each 5 mL solution contains 15.2 mg tamoxifen citrate, equivalent to 10 mg tamoxifen).
(a) Cancer of the breast
(i) Metastatic breast cancer
For the treatment of metastatic breast cancer in women, the usual dosage of tamoxifen is 20–40 mg daily, typically starting with the 20 mg dose. Dosages exceeding 20 mg daily are typically given in divided doses (morning and evening). A 20 mg oral dose is administered as 10 mL of the oral solution.
(ii) Adjuvant therapy of breast cancer
When tamoxifen is used as an adjunct to surgery and radiation therapy in the treatment of breast cancer, the usual dosage of the drug is 20–40 mg daily. Dosages exceeding 20 mg daily typically are given in divided doses (morning and evening). The optimum duration of adjuvant tamoxifen therapy has not been established, but therapy for about 5 years has become the norm.
When tamoxifen is used in combination with chemotherapy as an adjunct to surgery in the treatment of breast cancer in postmenopausal women or in women 50 years of age or older who have positive axillary lymph nodes, the usual dosage of the drug is 10 mg twice daily. The optimum duration of adjuvant tamoxifen therapy has not been established.
(iii) Ductal carcinoma in situ (DCIS)
In women with DCIS following breast surgery and radiation therapy, tamoxifen is used for a recommended duration of 5 years to reduce the risk of invasive breast cancer.
(iv) Chemoprevention in women at high risk of breast cancer
For reduction in the incidence of breast cancer in women at high risk, the recommended dosage of tamoxifen is 20 mg daily given for 5 years.
(v) Male breast cancer
For the treatment of advanced (metastatic) breast cancer in men, the usual dosage of tamoxifen is 20–40 mg daily. Tamoxifen alone or in combination with radiation therapy was also used as an adjunct to surgery in the treatment of breast cancer in men at a dosage of 20 mg daily, usually for 1–2 years.
(b) Other uses
For anovulatory infertility, 20 mg of tamoxifen is administered daily on Days 2, 3, 4, and 5 of the cycle; if necessary, the daily dose may be increased to 40 mg and then 80 mg for subsequent courses; if menstrual cycles are irregular, the initial course may be started on any day, with subsequent course starting 45 days later or on Day 2 of cycle if menstruation occurs.
1.2.3. Trends in use
Although tamoxifen citrate is still available as a breast cancer treatment, it has largely been replaced by other treatments (AstraZeneca PLC, 2004, 2007).
2. Cancer in Humans
2.1. Cancer of the endometrium
In the sections that follow, the most important studies considered in the previous IARC Monograph (IARC, 1996) are included. Case reports are not considered.
Detection bias may pertain to both cohort studies and randomized clinical trials as tamoxifen is known to increase the frequency of symptoms such as vaginal bleeding or discharge, which may lead to gynaecological evaluation. In addition, tamoxifen is known to induce benign gynaecological changes such as endometrial hyperplasia and polyps. Other changes include poorly defined thickening of the endometrium that may be revealed by ultrasound examination.
The longer survival of tamoxifen-treated patients may lead to greater duration of follow-up in which second cancers may occur. The appropriate methods of statistical analysis in this context are life table analyses or analyses of rates based on person–years at risk.
2.1.1. Cohort studies
Of 11 cohort studies of women with breast cancer, three were based on data from the SEER (Surveillance, Epidemiology and End Results) Program in the United States of America (Curtis et al., 1996, 2004; Newcomb et al., 1999), all included substantial numbers of cases of endometrial cancer, and all found significant elevations of risk for endometrial cancer. Specific histological types were evaluated in the Curtis et al. (2004) study. The relative risk was higher for malignant mixed mullerian tumours (MMMTs) than for endometrial adenocarcinomas, although the excess absolute risk was smaller—an additional 1.4 versus 8.4 cancers per 10000 women per year, respectively. [The Working Group noted that the Curtis et al. (2004) study is an extension of Curtis et al. (1996). It is probable that there is some overlap with the cohort reported by Newcomb et al. (1999), but the authors do not discuss this nor cite the paper by Newcomb et al. (1999). In these studies, the absence of hysterectomy could not be confirmed, nor were individual records of tamoxifen use available. Misclassification of hormonal treatment in the studies may have led to an underestimation of the difference in risk for cancer of the uterine corpus between the groups.]
Of the remaining cohort studies of women with breast cancer, one was a nested case–control study (Cook et al., 1995) in which tamoxifen use was more common in the controls (31% versus 26%). Another small cohort study (Katase et al., 1998) found no increase in risk of endometrial cancer in those treated with tamoxifen. Three of the other cohort studies found a positive but non-significant elevation of the risk of endometrial cancer (Matsuyama et al., 2000; Ursic-Vrscaj et al., 2001; Yamazawa et al., 2006), though the numbers of endometrial cancer cases in these studies were small. Of the two remaining cohort studies, one (Bouchardy et al., 2002) found significantly elevated risks of uterine cancer with tamoxifen use, and the other (Lavie et al., 2008), borderline increases in risk. There was one additional cohort study, based on women at high risk of breast cancer, with a significantly increased risk of endometrial cancer following tamoxifen use (Beiner et al., 2007).
See Table 2.1 available online at http://monographs.iarc.fr/ENG/Monographs/vol100A/100A-08-Table2.1.pdf
2.1.2. Case–control studies
The case–control studies considered by the Working Group were those that compared tamoxifen use in women with breast cancer who did (cases) or did not (controls) subsequently develop endometrial cancer. A fundamental requirement for the controls is that they were at risk of developing endometrial cancer (i.e. they had an intact uterus). Determinants of risk for endometrial cancer are confounding factors in the studies discussed below only to the extent that they influence the likelihood of tamoxifen prescription. As in any case–control study, information and selection bias may also be present. Finally, the possibility that endometrial cancer was diagnosed preferentially in women who had received tamoxifen constitutes a potential bias that is considered in the introductory remarks to Section 2.1.
All of the seven case–control studies found elevations in risk of endometrial cancer following tamoxifen use. Three of the studies found significant elevations of risk following tamoxifen use (Mignotte et al., 1998; Bergman et al., 2000; Swerdlow & Jones, 2005). In two (Bergman et al., 2000; Swerdlow & Jones, 2005), greater (significant) increases in risk followed durations of use of tamoxifen of 5 years or more. The study of Chu et al. (2007) was conducted to examine whether a genetic variant of the CYP3A4 gene, CYP3A4*1B, influences endometrial cancer risk–alone or when associated with tamoxifen exposure, as tamoxifen is metabolized by various cytochrome P450 (CYP) enzymes, but predominantly by CYP3A4. This resulted in the finding of an increase in risk in women who carried the CYP3A4*1B allele following treatment with tamoxifen.
[Although several potential confounders were not systematically addressed in most of the case–control studies, the Working Group considered that these were unlikely to have had a major effect on the reported relative risks.]
See Table 2.2 available online at http://monographs.iarc.fr/ENG/Monographs/vol100A/100A-08-Table2.2.pdf
2.1.3. Randomized trials
In most of the randomized trials of breast cancer treatment, small numbers of endometrial cancers were reported. In two of the largest trials, however, there was a strong and statistically significant association between risk of endometrial cancer and use of tamoxifen (Fisher et al., 1996; Rutqvist & Johansson, 2007). Data from the large majority of the trials individually cited in IARC (1996) were included in the meta-analyses described below. Therefore, these trials are not further discussed here.
All of the reports show statistically significant elevations of the risk of endometrial cancer except in the trial of Delozier et al. (2000), in which all participants received tamoxifen for at least 2 years. The pooled analysis of The Early Breast Cancer Trialists’ Collaborative Group (1998) was the largest of these analyses. Information was collected in 1995, which was analysed centrally. The incidence of endometrial cancer had approximately quadrupled in trials of 5 years of tamoxifen use (although the number of cases was small, and these ratios were not significantly different from the findings at 2 years). Mortality from endometrial cancer was also significantly increased in those who received tamoxifen, 27 versus 5 deaths in controls, for a 10-year risk per 1000 of 1.7 versus 0.4. Two of the reports (Braithwaite et al., 2003; Cuzick et al., 2003) included findings from chemoprevention trials using tamoxifen as well as treatment trials. [The Working Group noted that there is some overlap in these reports with some of the treatment trial data included in The Early Breast Cancer Trialists’ Collaborative Group (1998) overview analysis, but the extent is unknown.] The 7-year follow-up data from the National Surgical Adjuvant Breast and Bowel Project chemoprevention trial confirmed the earlier reported excess of endometrial cancer in those who received tamoxifen (20 mg/day) (Fisher et al., 2005).
See Table 2.3 available online at http://monographs.iarc.fr/ENG/Monographs/vol100A/100A-08-Table2.3.pdf
2.2. Contralateral breast cancer
Although for some of the smaller trials of breast cancer treatment there seemed to be little difference in the number of contralateral breast cancer in tamoxifen-treated women compared with controls, for the larger trials, there was a substantially and significantly reduced risk for contralateral breast cancer in tamoxifen-treated women compared with controls. In a combined analysis of nearly all trials published in 1992 with data available to 1990, there was a significant reduction of 39% in contralateral breast cancer in the tamoxifen-treated groups (IARC, 1996), confirmed by subsequent overview analyses. In the Cuzick et al. (2003) overview analysis of nine treatment trials, the benefit was restricted to reduction in estrogen-receptor-positive cancers.
See Table 2.4 available online at http://monographs.iarc.fr/ENG/Monographs/vol100A/100A-08-Table2.4.pdf
Four cohort studies reported on contralateral breast cancer. In three (Cook et al., 1995; Curtis et al., 1996; Newcomb et al., 1999), the risk of contralateral breast cancer was reduced in the tamoxifen-treated women, compared with women with no reported tamoxifen use. However, Matsuyama et al. (2000) reported no risk reduction in their cohort study from Japan.
See Table 2.5 available online at http://monographs.iarc.fr/ENG/Monographs/vol100A/100-A-08-Table2.5.pdf
2.3. Chemoprevention of cancer of the breast
Four trials of chemoprevention have been conducted with one being stopped because the desired end-point was reached earlier than anticipated (Fisher et al., 2005). The latest data available from this trial are summarized in Table 2.6 (available online at http://monographs.iarc.fr/ENG/Monographs/vol100A/100A-08-Table2.6.pdf), together with the results of a meta-analysis combining earlier data from this trial with that available from the three other trials (Cuzick et al., 2003). It is apparent that there is a significant reduction in the incidence of breast cancer in the women who received tamoxifen. As for contralateral breast cancer, the benefit was restricted to reduction in estrogen-receptor-positive cancers.
2.4. Gastrointestinal cancers
Although an excess of gastrointestinal cancer was reported following an early combined analysis of three Scandinavian trials, this was not confirmed by other studies reported to 1996 (IARC, 1996). Data on gastrointestinal cancer was reported in five cohort studies (Curtis et al., 1996; Newcomb et al., 1999; Matsuyama et al., 2000; Srinivasan et al., 2005; Chandanos et al., 2006), with only one providing data on all gastrointestinal sites combined (Curtis et al., 1996). Three studies provided data on oesophageal cancer, four on stomach cancer, and four on colon or colorectal cancer. None of the studies reported significant excess risks for oesophageal, stomach or colorectal cancer in comparisons between breast cancer patients treated with tamoxifen and those who did not receive tamoxifen. However, Newcomb et al. (1999) reported a borderline significant positive association between hormonal therapy use and colorectal cancer in the period 5 or more years after diagnosis, while Matsuyama et al. (2000) and Chandanos et al. (2006) reported significant excesses of stomach cancer in comparison to the general population. [The Working Group thought that such comparisons are biased, and although comparisons between subjects with breast cancer treated and not treated with tamoxifen may also not be entirely valid as discussed above, they are preferable to comparisons with the general population.]
In The Early Breast Cancer Trialists’ Collaborative Group (1998) analysis, tamoxifen had no apparent effect on the incidence of colorectal cancer. However, in the Braithwaite et al. (2003) meta-analysis, tamoxifen was associated with significantly increased risks of gastrointestinal cancers (reported in 16 trials), with a relative risk of 1.31 (95% CI: 1.01–1.69).
See Table 2.7 available online at http://monographs.iarc.fr/ENG/Monographs/vol100A/100A-08-Table2.7.pdf
2.5. Cancer of the ovary
Five cohort studies (Cook et al., 1995; Curtis et al., 1996; Newcomb et al., 1999; Matsuyama et al., 2000; Ursic Vrscaj et al., 2001), and two case–control studies (Metcalfe et al., 2005; Swerdlow & Jones, 2007) evaluated the role of tamoxifen therapy in women with breast cancer in relation to the risk of subsequent ovarian cancer. No study showed any indications of increased risk, though the numbers of cases in some of the cohort studies were very small.
See Table 2.8 available online at http://monographs.iarc.fr/ENG/Monographs/vol100A/100A-08-Table2.8.pdf and Table 2.9 available online at http://monographs.iarc.fr/ENG/Monographs/vol100A/100A-08-Table2.9.pdf
2.6. Synthesis
In summary, the potential effect of tamoxifen in increasing the risk of endometrial cancer among women with breast cancer has been reported in nine cohort studies, four case–control studies, five randomized controlled treatment trials, and one major chemoprevention trial; the majority of these in meta-analyses, though there were four separate reports from individual trials. The data from the observational studies and randomized controlled trials are largely consistent in showing that tamoxifen, whether given as adjuvant therapy for women with breast cancer or for chemoprevention in women at high risk for breast cancer, increases the risk for endometrial cancer. Also, there is evidence that the use of tamoxifen in the treatment of breast cancer significantly reduces the incidence of contralateral breast cancer. The use of tamoxifen, when given for chemoprevention, reduces the incidence of estrogen-receptor-positive breast cancers.
Finally, there is some indication that the risk of various forms of gastrointestinal cancer may be increased in tamoxifen-treated patients, however the data are not consistent.
3. Cancer in Experimental Animals
Tamoxifen has been tested for carcinogenicity by oral and subcutaneous administration to adult and infant mice and rats, and by transplacental exposure to mice.
3.1. Oral administration
3.1.1. Mouse
In mice treated orally, tamoxifen increased the incidence of benign ovarian and benign testicular tumours in one study (Tucker et al., 1984), but did not increase the tumour incidence in two other studies (Carthew et al., 1996a; Martin et al., 1997).
3.1.2. Rat
Rats dosed orally with tamoxifen had an increased incidence of benign and malignant liver cell tumours in multiple studies (Greaves et al., 1993; Hard et al., 1993; Hirsimäki et al., 1993; Williams et al., 1993, 1997; Ahotupa et al., 1994; Hasmann et al., 1994; Carthew et al., 1995b; Dragan et al., 1995; Kärki et al., 2000; Carthew et al., 2001; Kasahara et al., 2003). When given at a lower dose level, tamoxifen decreased the incidence of benign and malignant mammary gland tumours in female rats, and benign pituitary tumours in both sexes (Maltoni et al., 1997).
See Table 3.1.
Table 3.1
Studies of cancer in experimental animals exposed to tamoxifen (oral exposure).
3.2. Subcutaneous administration
3.2.1. Mouse
In mice treated subcutaneously, tamoxifen decreased the incidence of mammary tumours in multiple studies (Jordan et al., 1990, 1991). One study using a transgenic mouse model susceptible to spontaneous mammary tumours resulted in an increased incidence of malignant mammary tumours (Jones et al., 2005).
3.2.2. Rats
Female rats administered the tamoxifen metabolite 4-hydroxytamoxifen subcutaneously had a decreased incidence of benign and malignant mammary tumours in one study (Sauvez et al., 1999).
See Table 3.2.
Table 3.2
Studies of cancer in experimental animals exposed to tamoxifen (subcutaneous administration).
3.3. Perinatal administration
3.3.1. Mouse
Tamoxifen given orally or subcutaneously to female neonatal mice increased the incidence of uterine tumours in one study (without an obvious dose–response) (Newbold et al., 1997), but had no effect upon urogenital tumours in three other studies (Green et al., 2005; Waalkes et al., 2006a; Razvi et al., 2007). Male mice exposed transplacentally to arsenite and treated neonatally with tamoxifen had a decreased incidence of benign and malignant lung tumours in one study (Waalkes et al., 2006b). One study in which tamoxifen was administered transplacentally to mice was negative (Diwan et al., 1997).
3.3.2. Rat
The administration of tamoxifen to female neonatal rats caused an increase in reproductive tract tumours in one study (Carthew et al., 2000), but no effect in another study of shorter duration with fewer animals (Karlsson, 2006).
See Table 3.3.
Table 3.3
Studies of cancer in experimental animals exposed to tamoxifen (perinatal exposure).
3.4. Administration with known carcinogens
In several studies in both male and female rats, tamoxifen enhanced the hepatocarcinogenicity of previously administered N,N-diethylnitrosamine (IARC, 1996). In one study in rats, tamoxifen enhanced the development of N-nitrosodiethylamine-induced kidney tumours (Dragan et al., 1995). In another study, the administration of tamoxifen to pregnant rats increased mammary gland tumours in offspring subsequently treated with 7,12-dimethylbenz[a]anthracene (Halakivi-Clarke et al., 2000).
See Table 3.4.
Table 3.4
Studies of cancer in experimental animals exposed to tamoxifen and known carcinogens.
3.5. Synthesis
Oral administration of tamoxifen increased the incidence of testicular tumours in one study in mice and malignant liver cell tumours in multiple studies in rats. At lower dose level, tamoxifen decreased the incidence of benign and malignant mammary gland tumours in female rats, and pituitary tumours in both sexes.
Subcutaneous administration of tamoxifen decreased the incidence of mammary tumours in multiple studies in mice. One study using a transgenic mouse model showed an increased incidence in mammary tumours.
Perinatal exposure to tamoxifen increased the incidence of reproductive tract tumours in mice and rats.
4. Other Relevant Data
In the previous IARC Monograph (IARC, 1996), tamoxifen was found to increase liver tumour incidence in rats. The available evidence indicated that tamoxifen is both a genotoxic carcinogen and a tumour promoter in rat liver, and that humans are likely to be less susceptible to the genotoxicity of the drug. It was suggested that tissue-specific effects of tamoxifen binding to the estrogen receptor on gene expression might be involved in the ability of tamoxifen to increase or decrease tumour risk. The pertinent mechanistic data that appeared since this review are summarized below.
4.1. Absorption, distribution, metabolism, and excretion
(a) Humans
Tamoxifen is well absorbed after oral administration, and appears to be more than 99% bound to plasma proteins (mostly to albumin) (Lien et al., 1989). The absorption of tamoxifen shows a wide interindividual variation, which is probably due to differences in liver metabolism and differences in absorption in the gastrointestinal tract. In rats, mice, dogs and rhesus monkeys, most of the dosed material appears in the faeces, but bile-duct cannulation experiments with dogs and rats showed also a biliary excretion (Fromson et al., 1973a).
The pharmacokinetics of tamoxifen appear to be biphasic, with a distribution phase of 7–14 hours, and an elimination phase of about 7 days (Fromson et al., 1973b). The elimination half-life of N-desmethyltamoxifen is around 7 days, and 4-hydroxytamoxifen has a shorter half-life than tamoxifen (Buckley & Goa, 1989).
Several metabolites have been identified in the urine and plasma of human breast cancer patients (IARC, 1996). Metabolites detected in plasma include tamoxifen, N-desmethyltamoxifen, and tamoxifen-N-oxide; and in urine, glucuronides of four hydroxylated metabolites (4-hydroxytamoxifen, 4-hydroxy-N-desmethyltamoxifen, dihydroxytamoxifen and another monohydroxy- (possibly α-hydroxy) N-desmethyltamoxifen) (Poon et al., 1993). In another study, seven metabolites were identified in plasma (N-didesmethyltamoxifen, α-hydroxytamoxifen, 4-hydroxytamoxifen, tamoxifen-N-oxide, α-hydroxy-N-desmethyltamoxifen, 4-hydroxy-N-desmethyltamoxifen, and 4-hydroxytamoxifen-N-oxide) (Poon et al., 1995).
In biopsy and autopsy samples obtained from patients with breast cancer treated with tamoxifen, levels of tamoxifen and its metabolites (N-desmethyl-, N-didesmethyl-, 4-hydroxy- and 4-hydroxy-N-desmethyl-) were 10- to 60-fold higher in tissues (liver, lung, pancreas, brain, adipose) than in serum, with particularly high levels in liver and lung tissues. Similarly, pancreatic tumours, and brain metastases from breast cancer were found to contain high levels of the drug. Specimen of skin and bone tissue also contained tamoxifen and some metabolites (Lien et al., 1991). Furthermore, tamoxifen, 4-hydroxytamoxifen and N-desmethyltamoxifen were also found in postmortem and biopsy analyses of liver from tamoxifen-treated patients (Martin et al., 1995).
CYP3A4 and, to a lesser extent, isoforms 2D6, 2B6, 3A5, 2C9, and 2C19 mediate the conversion of tamoxifen to α-hydroxytamoxifen (Notley et al., 2005), whereas hydroxysteroid sulfotransferase 2A1 (SULT2A1) catalyses the formation of sulfate ester from α-hydroxytamoxifen (Apak & Duffel, 2004). A recent study (Singh et al., 2008) demonstrated the expression of genes encoding the enzymes CYP3A4 and SULT2A1 involved in the bioactivation of tamoxifen in the human endometrium. It has been reported that women carrying CYP3A4*1B, a variant of CYP3A4, are at increased risk for tamoxifen-induced endometrial cancer (Chu et al., 2007).
(b) Experimental systems
In experimental animals, concentrations of tamoxifen and its metabolites are 8- to 70-fold higher in tissues (brain, adipose, liver, heart, lung, kidney, uterus, testis) than in serum. The highest levels are found in lung and liver tissue, with substantial amounts found in kidney and adipose tissue (Lien et al., 1991).
Tamoxifen can be metabolized in vitro by both microsomal cytochrome P450 and flavin mono-oxygenase pathways to intermediates that bind irreversibly to microsomal proteins (Mani & Kupfer, 1991). Incubation of tamoxifen with rat liver microsomes results in the formation of three major polar metabolites (N-oxide, N-desmethyl and 4-hydroxy derivatives) (Mani et al., 1993, 1994). Peroxidases may also metabolize tamoxifen to a reactive intermediate that binds covalently to protein (Davies et al., 1995), and to DNA (Pathak et al., 1995).
In both human liver homogenate and human hepatic G2 cell line treated with a mixture of tamoxifen and its deuterated analogues, the following metabolites were detected: α-hydroxytamoxifen, 4-hydroxytamoxifen, N-desmethyltamoxifen, and tamoxifen N-oxide. In the liver homogenate, N-didesmethyltamoxifen was also detected (Poon et al., 1995).
When primary cultures of human, rat and mouse hepatocytes were incubated with tamoxifen (10 μM) for 18–24 hours, the concentration of α-hydroxytamoxifen in the medium was 50-fold lower in the human cultures than in the rat and mouse cultures (Phillips et al., 1996a).
4.2. Genetic and related effects
4.2.1. Direct genotoxicity
(a) DNA adducts
(i) Humans
Tamoxifen–DNA adducts have not been detected in human liver in vivo (IARC, 1996), and the low level of DNA covalent-binding by α-hydroxytamoxifen in cultured human hepatocytes (Phillips et al., 1996a) probably reflects the intrinsic chemical reactivity of α-hydroxytamoxifen rather than enzymatic activation, as this metabolite is a poor substrate for human sulfotransferases (Glatt et al., 1998a; Shibutani et al., 1998a). Moreover, the glucuronidation pathway predominates in incubations of α-hydroxytamoxifen with human liver microsomes (Boocock et al., 2000), which presumably leads to detoxification.
There are conflicting results on the formation of tamoxifen–DNA adducts in humans. The evidence for (Hemminki et al., 1996; Shibutani et al., 1999, 2000a; Martin et al., 2003) and against (Carmichael et al., 1996, 1999; Beland et al., 2004a) such adducts in the human endometrium in vivo has been reported by several groups. This is also the case in studies on the formation of tamoxifen–DNA adducts from incubation of tamoxifen with human endometrium explants, with positive (Sharma et al., 2003) and negative (Beland et al., 2004b) findings being reported in samples from the same origin. Some studies reported the presence of such adducts in white blood cells from tamoxifen-treated patients (Hemminki et al., 1997; Umemoto et al., 2004), while others reported negative results (Phillips et al., 1996b; Bartsch et al., 2000). With the exception of the study by Martin et al. (2003), who used accelerator mass spectrometry, most studies used 32P-postlabelling for adduct detection. Beland et al. (2004a, b) used HPLC coupled with tandem mass spectrometry, which can provide unequivocal structural characterization.
A recent study has reported the presence of (E)-α-(deoxyguanosin-N2-yl)tamoxifen (dG-Tam) at levels of 1–7 adducts/109 nucleotides in enzymatically hydrolysed colorectal DNA from 3/10 women administered a single dose of 20 mg 14C-labelled tamoxifen approximately 18 hours before colon resection surgery. The detection methodology involved HPLC coupled with accelerator mass spectrometry, and the identification was based upon comparison with an authentic adduct standard. All colon samples had detectable levels of CYP3A4 (Brown et al., 2007).
(ii) Experimental systems
DNA adducts have been detected at dose-dependent levels in rat liver following administration of tamoxifen (IARC, 1996), and some of its metabolites, such as N-desmethyltamoxifen, α-hydroxytamoxifen, and α-hydroxy-N-desmethyltamoxifen (Brown et al., 1998, 1999; Martin et al., 1998; Phillips et al., 1999, 2005; Gamboa da Costa et al., 2000, 2001; White et al., 2001). A quantitatively minor phase I pathway that leads to the metabolic activation of tamoxifen to DNA-binding electrophiles in rat liver is catalysed by CYP3A enzymes. This involves hydroxylation at the allylic (α) carbon of tamoxifen (Kim et al., 2003) and N-desmethyltamoxifen, which is then followed by phase II conjugation. Although acetyltransferases have been proposed as mediators in the activation of α-hydroxylated tamoxifen metabolites, the most convincing evidence indicates that activation occurs through sulfotransferase-mediated sulfation, specifically by the STA2 isoform of hydroxysteroid sulfotransferase (Davis et al., 1998, 2000; Glatt et al., 1998a, b; Shibutani et al., 1998a, b; Kim et al., 2005; Phillips et al., 2005). In addition, a parallel adduct formation pathway involving N-demethylation, as well as α-hydroxylation and O-sulfonation occurs (Fig. 4.1). The N-demethylation of tamoxifen is also mediated by the CYP3A subfamily (IARC, 1996). In-vitro reactions conducted with either the synthetic model esters, α-acetoxytamoxifen and α-acetoxy-N-desmethyltamoxifen (Osborne et al., 1996; Dasaradhi & Shibutani, 1997; Kitagawa et al., 2000) or the corresponding synthetic sulfates (Dasaradhi & Shibutani, 1997; Gamboa da Costa et al., 2000) have led to the identification of the major DNA adducts as (E)-α-(deoxyguanosin-N2-yl)tamoxifen (dG-Tam) and (E)-α-(deoxyguanosin-N2-yl)-N-desmethyltamoxifen (dG-desMe-Tam), which exist as mixtures of epimers at the allylic carbon. Minor adducts from these reactions include the Z diastereomers from dG-Tam and dG-desMe-Tam (Dasaradhi & Shibutani, 1997; Osborne et al., 1997; Kitagawa et al., 2000), and a deoxyadenosine-tamoxifen adduct, linked through the amino group of adenine (Osborne et al., 1997). Comparison with characterized synthetic standards has confirmed that dG-Tam and dG-desMeTam are the major adducts formed in rat liver following treatment with tamoxifen regardless of the rat strain, the route of administration, or the length of exposure (Osborne et al., 1996; Rajaniemi et al., 1998, 1999; Brown et al., 1999; Phillips et al., 1999; Firozi et al., 2000; Gamboa da Costa et al., 2000). Interestingly, the R enantiomers of α-hydroxytamoxifen (Osborne et al., 2001) and α-hydroxy-N-desmethyltamoxifen (Osborne et al., 2004) have much higher binding affinity in rat hepatocytes than the corresponding S isomers, presumably as a result of better affinity of the R enantiomers for the sulfotransferases.

Figure 4.1
Proposed pathways of activation of tamoxifen in rat liver.
Although a significant level of the didesmethylated analogue of dG-Tam and dG-desMeTam was detected in rat liver following administration of the putative metabolite, α-hydroxy-N,N-didesmethyltamoxifen, the low extent of binding obtained upon dosage with N,N-didesmethyltamoxifen indicates that metabolic activation to α-hydroxy-N,N-didesmethyltamoxifen is a minor pathway in the rat (Gamboa da Costa et al., 2003). Likewise, metabolism via 4-hydroxytamoxifen does not seem to be a significant pathway to DNA-adduct formation in the rat (Beland et al., 1999; Osborne et al., 1999; Kim et al., 2006a), despite the fact that the metabolite can be activated enzymatically to products covalently bound to DNA in cell-free or subcellular systems (Pathak et al., 1995, 1996), and both its oxidation products, 4-hydroxytamoxifen quinone methide (Marques & Beland, 1997) and α-4-dihydroxytamoxifen (Hardcastle et al., 1998) give DNA adducts upon reaction with DNA in vitro. An additional minor pathway to DNA adduct formation in rat liver has been reported to proceed via α-hydroxytamoxifen N-oxide, again involving binding at the α carbon through the exocyclic nitrogen of deoxyguanosine (Umemoto et al., 1999, 2001).
While tamoxifen–DNA adducts are consistently detected in rat liver, most studies have not detected DNA adducts in the uterus and other extrahepatic tissues from rats administered tamoxifen or tamoxifen metabolites (Li et al., 1997; Brown et al., 1998; Beland et al., 1999; Carthew et al., 2000; Gamboa da Costa et al., 2001; Phillips et al., 2005). However, one study, which involved the use of accelerator mass spectrometry, reported that [14C]tamoxifen binds to DNA in the liver, intestine, reproductive tract, spleen, lung, and kidney of rats dosed orally (White et al., 1997). However, this methodology does not provide any structural information. [The Working Group noted that it was not clear whether the measured radioactivity corresponded exclusively to tamoxifen covalently bound to DNA.]
Tamoxifen also forms DNA adducts in mouse liver, though the levels are typically lower than in the rat (IARC, 1996). In addition, chronic exposure does not lead to accumulation of DNA adducts, which, combined with the absence of tamoxifen-induced cell proliferation, may account for the lack of hepatic carcinogenicity in the mouse, as opposed to the rat (Martin et al., 1997). Similarly to what is found in the rat, the major DNA adducts in mouse liver are dG-Tam and dG-desMeTam; although still minor, the adduct diastereomers derived from α-hydroxytamoxifen N-oxide make up a higher proportion in the mouse than in the rat (Umemoto et al., 2000, 2001). The presence of DNA adducts in mouse extrahepatic tissues, including the uterus, has not been investigated.
Low levels of combined dG-Tam and dG-desMeTam were detected by different methods in the liver, brain cortex, kidney, ovary, and uterus of a group of three female cynomolgus monkeys dosed with a daily regimen of 2 mg tamoxifen/kg body weight for 30 days (Schild et al., 2003; Shibutani et al., 2003). These studies have shown that tamoxifen DNA adducts can be formed in extrahepatic tissues of non-human primates.
(b) Additional genotoxic effects
Tamoxifen induces micronuclei in metabolically proficient human cells and causes aneuploidy and chromosomal aberrations in rat liver (IARC, 1996; Styles et al., 1997). Moreover, both tamoxifen and α-hydroxytamoxifen cause mutations in the lacI reporter gene and the cII gene in the livers of Big Blue transgenic rats (Davies et al., 1996, 1997, 1999; Styles et al., 2001; Chen et al., 2002; Gamboa da Costa et al., 2002) although α-hydroxytamoxifen causes significantly higher mutant frequencies than does tamoxifen, the mutational spectra induced by the two compounds are very similar in both genes, with the predominant mutations being G→T transversions. Mutations are not observed in extrahepatic tissues, including the uterus, which is in agreement with the general lack of detection of DNA adducts in rat tissues other than the liver. Consistent with the mutation profile in rat liver, when single-stranded shuttle vectors containing each of the four dG-Tam diastereomers were transfected into simian kidney (COS7) cells, the prevalent mutations were, in all instances, G→T transversions (Terashima et al., 1999). Likewise, when α-hydroxytamoxifen was tested in V79-rHSTa cells, a mammalian cell line with stable expression of rat hydroxysteroid sulfotransferase A (STA2), mutations at the Hprt gene were mainly GC→TA transversions, although single G:C base-pair deletions and partial/complete exon skippings were also observed, almost exclusively at guanines on the non-transcribed strand (Yadollahi-Farsani et al., 2002). Additionally, both 4-hydroxytamoxifen quinone methide and the model ester, α-acetoxytamoxifen, are promutagenic using adducted pSP189 plasmid DNA containing the supF gene transfected into cultured human fibroblasts and kidney cells (McLuckie et al., 2002, 2005). Experiments involving the use of site-specific modified oligonucleotides as templates in primer extension reactions with several mammalian DNA polymerases indicate that all four dG-Tam diastereomers have high miscoding potential (G→T mutation) (Shibutani & Dasaradhi, 1997; Yasui et al., 2006). These adducts undergo nucleotide excision repair in vitro (Shibutani et al., 2000b). A comparative study in excision-repair-deficient (XPC knockout) and wild-type mice indicated that they have similar removal rates in both strains, which indicates that hepatic tamoxifen DNA-adducts are not efficiently repaired by this pathway (Kim et al., 2006b).
A study of the DNA-damaging potential of tamoxifen in normal human peripheral blood lymphocytes and MCF-7 breast cancer cells using the comet assay reported evidence for the presence of free radicals, which might account, in part, for the genotoxicity of tamoxifen under the experimental conditions presumably due to incomplete repair of double-strand breaks (Wozniak et al., 2007).
Both tamoxifen and its β-chlorinated analogue toremifene, which has a much lower potential for DNA-adduct formation (Gamboa da Costa et al., 2007), are associated with endometrial K-ras codon 12 mutations (Wallén et al., 2005), although a different study concluded that toremifene has a much lower potential than tamoxifen for K-ras mutation induction in the human endometrium (Hachisuga et al., 2005). [The Working Group noted that mutations in TP53 and K-RAS are low-frequency lesions in the common form of endometrial cancer and even those mutations appear late in the course of tumour development (Sherman, 2000).]
4.2.2. Estrogen-receptor-mediated mechanism
Experimental evidence increasingly supports the importance of estrogen-receptor-mediated gene regulation as the mechanism responsible for the differential action of tamoxifen in distinct tissues (Wu et al., 2005). Selective estrogen-receptor modulators such as tamoxifen have tissue-specific estrogenic activity. Tamoxifen is an estrogen-receptor antagonist in the breast but an estrogen-receptor agonist in the bone and uterus. The two main forms of the estrogen receptor, estrogen receptor-α and estrogen receptor-β, have different tissue expression profiles. The uterus predominantly expresses estrogen receptor-α but the observation of increased cell proliferation and excessive response to estrogen in estrogen-receptor-β-knockout mice has suggested that estrogen receptor-β could modulate estrogen receptor-α in the uterus, and have an antiproliferative role (Lecce et al., 2001). Tamoxifen stimulates proliferation of the human endometrial epithelium (Mourits et al., 2002). Tamoxifen-liganded estrogen receptors associate with multiple co-activator proteins, which together determine tamoxifen binding and transactivation activity (Shang, 2006). Tamoxifen regulates gene transcription in epithelial cells from type I endometrial carcinomas (Wu et al., 2005), and transcriptional responses have been identified in epithelial cells but not in stromal cells (Pole et al., 2004). There is also evidence that the genes targeted by tamoxifen differ from those targeted by estrogen (Pole et al., 2005).
4.3. Synthesis
There is strong evidence that in rat liver, tamoxifen is a genotoxic carcinogen through a pathway involving α-hydroxylation, sulfation of the α-hydroxy metabolite, and subsequent DNA-adduct formation.
Evidence for the role of this pathway in induction of human endometrial tumours is less compelling; rather, the data suggest that the carcinogenicity of tamoxifen is associated with an estrogen-receptor-dependent pathway.
5. Evaluation
There is sufficient evidence in humans for the carcinogenicity of tamoxifen. Tamoxifen causes cancer of the endometrium.
For cancer of the female breast, there is evidence suggesting lack of carcinogenicity. An inverse relationship has been established between exposure to tamoxifen and cancer of the female breast.
There is sufficient evidence in experimental animals for the carcinogenicity of tamoxifen.
Tamoxifen is carcinogenic to humans (Group 1).
References
- Ahotupa M, Hirsimäki P, Pärssinen R, Mäntylä E. Alterations of drug metabolizing and antioxidant enzyme activities during tamoxifen-induced hepatocarcinogenesis in the rat. Carcinogenesis. 1994;15:863–868. [PubMed: 8200088] [CrossRef]
- Albain KS. Adjuvant chemotherapy for lymph node-negative, estrogen receptor-negative breast cancer: a tale of three trials. J Natl Cancer Inst. 2004;96:1801–1804. [PubMed: 15601631] [CrossRef]
- Apak TI, Duffel MW. Interactions of the stereoisomers of alpha-hydroxytamoxifen with human hydroxysteroid sulfotransferase SULT2A1 and rat hydroxysteroid sulfotransferase STa. Drug Metab Dispos. 2004;32:1501–1508. [PubMed: 15371299] [CrossRef]
- AstraZeneca Pharmaceuticals LP (2005). NOLVADEX® Tamoxifen Citrate, package insert. Wilmington, DE, 41 pp.
- AstraZeneca PLC (2004). Annual Report and Form 20-F Information 2003. London, 144 pp.
- AstraZeneca PLC (2007). Annual Report and Form 20-F Information 2006. London, 184 pp.
- Bartsch H, Phillips DH, Nair J, et al. Lack of evidence for tamoxifen- and toremifene-DNA adducts in lymphocytes of treated patients. Carcinogenesis. 2000;21:845–847. [PubMed: 10753226] [CrossRef]
- Beiner ME, Finch A, Rosen B, et al. Hereditary Ovarian Cancer Clinical Study Group. The risk of endometrial cancer in women with BRCA1 and BRCA2 mutations. A prospective study. Gynecol Oncol. 2007;104:7–10. [PubMed: 16962648] [CrossRef]
- Beland FA, Churchwell MI, Doerge DR, et al. Electrospray ionization-tandem mass spectrometry and 32P-postlabeling analyses of tamoxifen-DNA adducts in humans. J Natl Cancer Inst. 2004;96:1099–1104. a. [PubMed: 15265972] [CrossRef]
- Beland FA, Churchwell MI, Hewer A, et al. Analysis of tamoxifen-DNA adducts in endometrial explants by MS and 32P-postlabeling. Biochem Biophys Res Commun. 2004;320:297–302. b. [PubMed: 15219826] [CrossRef]
- Beland FA, McDaniel LP, Marques MM. Comparison of the DNA adducts formed by tamoxifen and 4-hydroxytamoxifen in vivo. Carcinogenesis. 1999;20:471–477. [PubMed: 10190564] [CrossRef]
- Bergman L, Beelen ML, Gallee MP, et al. Risk and prognosis of endometrial cancer after tamoxifen for breast cancer. Comprehensive Cancer Centres’ ALERT Group. Assessment of Liver and Endometrial cancer Risk following Tamoxifen. Lancet. 2000;356:881–887. [PubMed: 11036892] [CrossRef]
- Boocock DJ, Maggs JL, Brown K, et al. Major inter-species differences in the rates of O-sulphonation and O-glucuronylation of alpha-hydroxytamoxifen in vitro: a metabolic disparity protecting human liver from the formation of tamoxifen-DNA adducts. Carcinogenesis. 2000;21:1851–1858. [PubMed: 11023543] [CrossRef]
- Bouchardy C, Verkooijen HM, Fioretta G, et al. Increased risk of malignant mullerian tumor of the uterus among women with breast cancer treated by tamoxifen. J Clin Oncol. 2002;20:4403. [PubMed: 12409344] [CrossRef]
- Braithwaite RS, Chlebowski RT, Lau J, et al. Meta-analysis of vascular and neoplastic events associated with tamoxifen. J Gen Intern Med. 2003;18:937–947. [PMC free article: PMC1494944] [PubMed: 14687281] [CrossRef]
- Brown K, Brown JE, Martin EA, et al. Determination of DNA damage in F344 rats induced by geometric isomers of tamoxifen and analogues. Chem Res Toxicol. 1998;11:527–534. [PubMed: 9585484] [CrossRef]
- Brown K, Heydon RT, Jukes R, et al. Further characterization of the DNA adducts formed in rat liver after the administration of tamoxifen, N-desmethyltamoxifen or N, N-didesmethyltamoxifen. Carcinogenesis. 1999;20:2011–2016. [PubMed: 10506118] [CrossRef]
- Brown K, Tompkins EM, Boocock DJ, et al. Tamoxifen forms DNA adducts in human colon after administration of a single [14C]-labeled therapeutic dose. Cancer Res. 2007;67:6995–7002. [PubMed: 17638912] [CrossRef]
- Buckley MM, Goa KL. Tamoxifen. A reappraisal of its pharmacodynamic and pharmacokinetic properties, and therapeutic use. Drugs. 1989;37:451–490. [PubMed: 2661195] [CrossRef]
- Carmichael PL, Sardar S, Crooks N, et al. Lack of evidence from HPLC 32P-post-labelling for tamoxifen-DNA adducts in the human endometrium. Carcinogenesis. 1999;20:339–342. [PubMed: 10069474] [CrossRef]
- Carmichael PL, Ugwumadu AH, Neven P, et al. Lack of genotoxicity of tamoxifen in human endometrium. Cancer Res. 1996;56:1475–1479. [PubMed: 8603387]
- Carthew P, Edwards RE, Nolan BM, et al. Tamoxifen associated uterine pathology in rodents: relevance to women. Carcinogenesis. 1996;17:1577–1582. a. [PubMed: 8761412] [CrossRef]
- Carthew P, Edwards RE, Nolan BM, et al. Tamoxifen induces endometrial and vaginal cancer in rats in the absence of endometrial hyperplasia. Carcinogenesis. 2000;21:793–797. [PubMed: 10753217] [CrossRef]
- Carthew P, Lee PN, Edwards RE, et al. Cumulative exposure to tamoxifen: DNA adducts and liver cancer in the rat. Arch Toxicol. 2001;75:375–380. [PubMed: 11570696] [CrossRef]
- Carthew P, Martin EA, White IN, et al. Tamoxifen induces short-term cumulative DNA damage and liver tumors in rats: promotion by phenobarbital. Cancer Res. 1995;55:544–547. b. [PubMed: 7834623]
- Carthew P, Nolan BM, Edwards RE, Smith LL. The role of cell death and cell proliferation in the promotion of rat liver tumours by tamoxifen. Cancer Lett. 1996;106:163–169. b. [PubMed: 8844968] [CrossRef]
- Carthew P, Rich KJ, Martin EA, et al. DNA damage as assessed by 32P-postlabelling in three rat strains exposed to dietary tamoxifen: the relationship between cell proliferation and liver tumour formation. Carcinogenesis. 1995;16:1299–1304. a. [PubMed: 7788846] [CrossRef]
- Chandanos E, Lindblad M, Jia C, et al. Tamoxifen exposure and risk of oesophageal and gastric adenocarcinoma: a population-based cohort study of breast cancer patients in Sweden. Br J Cancer. 2006;95:118–122. [PMC free article: PMC2360495] [PubMed: 16755290] [CrossRef]
- Chen T, Gamboa da Costa G, Marques MM, et al. Mutations induced by alpha-hydroxytamoxifen in the lacI and cII genes of Big Blue transgenic rats. Carcinogenesis. 2002;23:1751–1757. [PubMed: 12376486] [CrossRef]
- Chu W, Fyles A, Sellers EM, et al. Association between CYP3A4 genotype and risk of endometrial cancer following tamoxifen use. Carcinogenesis. 2007;28:2139–2142. [PubMed: 17434921] [CrossRef]
- Cook LS, Weiss NS, Schwartz SM, et al. Population-based study of tamoxifen therapy and subsequent ovarian, endometrial, and breast cancers. J Natl Cancer Inst. 1995;87:1359–1364. [PubMed: 7658496] [CrossRef]
- Curtis RE, Boice JD Jr, Shriner DA, et al. Second cancers after adjuvant tamoxifen therapy for breast cancer. J Natl Cancer Inst. 1996;88:832–834. [PubMed: 8637050] [CrossRef]
- Curtis RE, Freedman DM, Sherman ME, Fraumeni JF Jr. Risk of malignant mixed mullerian tumors after tamoxifen therapy for breast cancer. J Natl Cancer Inst. 2004;96:70–74. [PubMed: 14709741] [CrossRef]
- Cuzick J, Powles T, Veronesi U, et al. Overview of the main outcomes in breast-cancer prevention trials. Lancet. 2003;361:296–300. [PubMed: 12559863] [CrossRef]
- Dasaradhi L, Shibutani S. Identification of tamoxifen-DNA adducts formed by alpha-sulfate tamoxifen and alpha-acetoxytamoxifen. Chem Res Toxicol. 1997;10:189–196. [PubMed: 9049430] [CrossRef]
- Davies AM, Martin EA, Jones RM, et al. Peroxidase activation of tamoxifen and toremifene resulting in DNA damage and covalently bound protein adducts. Carcinogenesis. 1995;16:539–545. [PubMed: 7697811] [CrossRef]
- Davies R, Gant TW, Smith LL, Styles JA. Tamoxifen induces G:C–>T:A mutations in the cII gene in the liver of lambda/lacI transgenic rats but not at 5′-CpG-3′ dinucleotide sequences as found in the lacI transgene. Carcinogenesis. 1999;20:1351–1356. [PubMed: 10383911] [CrossRef]
- Davies R, Oreffo VI, Bayliss S, et al. Mutational spectra of tamoxifen-induced mutations in the livers of lacI transgenic rats. Environ Mol Mutagen. 1996;28:430–433. [PubMed: 8991074] [CrossRef]
- Davies R, Oreffo VI, Martin EA, et al. Tamoxifen causes gene mutations in the livers of lambda/lacI transgenic rats. Cancer Res. 1997;57:1288–1293. [PubMed: 9102215]
- Davis W, Hewer A, Rajkowski KM, et al. Sex differences in the activation of tamoxifen to DNA binding species in rat liver in vivo and in rat hepatocytes in vitro: role of sulfotransferase induction. Cancer Res. 2000;60:2887–2891. [PubMed: 10850433]
- Davis W, Venitt S, Phillips DH. The metabolic activation of tamoxifen and alpha-hydroxytamoxifen to DNA-binding species in rat hepatocytes proceeds via sulphation. Carcinogenesis. 1998;19:861–866. [PubMed: 9635875] [CrossRef]
- Delozier T, Spielmann M, Macé-Lesec’h J, et al. Fédération Nationale des Centres de Lutte Contre le Cancer Breast Group. Tamoxifen adjuvant treatment duration in early breast cancer: initial results of a randomized study comparing short-term treatment with long-term treatment. J Clin Oncol. 2000;18:3507–3512. [PubMed: 11032592]
- Diwan BA, Anderson LM, Ward JM. Proliferative lesions of oviduct and uterus in CD-1 mice exposed prenatally to tamoxifen. Carcinogenesis. 1997;18:2009–2014. [PubMed: 9364013] [CrossRef]
- Dragan VP, Vaughan J, Jordan VC, Pitot HC. Comparison of the effects of tamoxifen and toremifene on liver and kidney tumor promotion in female rats. Carcinogenesis. 1995;16:2733–2741. [PubMed: 7586193] [CrossRef]
- Firozi PF, Vulimiri SV, Rajaniemi H, et al. Characterization of the major DNA adducts in the liver of rats chronically exposed to tamoxifen for 18 months. Chem Biol Interact. 2000;126:33–43. [PubMed: 10826652] [CrossRef]
- Fisher B, Costantino JP, Wickerham DL, et al. Tamoxifen for the prevention of breast cancer: current status of the National Surgical Adjuvant Breast and Bowel Project P-1 study. J Natl Cancer Inst. 2005;97:1652–1662. [PubMed: 16288118] [CrossRef]
- Fisher B, Dignam J, Bryant J, et al. Five versus more than five years of tamoxifen therapy for breast cancer patients with negative lymph nodes and estrogen receptor-positive tumors. J Natl Cancer Inst. 1996;88:1529–1542. [PubMed: 8901851] [CrossRef]
- Fromson JM, Pearson S, Bramah S. The metabolism of tamoxifen (I.C.I. 46,474). I. In laboratory animals. Xenobiotica. 1973;3:693–709. a. [PubMed: 4361333] [CrossRef]
- Fromson JM, Pearson S, Bramah S. The metabolism of tamoxifen (I.C.I. 46,474). II. In female patients. Xenobiotica. 1973;3:711–714. b. [PubMed: 4783632] [CrossRef]
- Gamboa da Costa G, Hamilton LP, Beland FA, Marques MM. Characterization of the major DNA adduct formed by alpha-hydroxy-N-desmethyltamoxifen in vitro and in vivo. Chem Res Toxicol. 2000;13:200–207. [PubMed: 10725117] [CrossRef]
- Gamboa da Costa G, Manjanatha MG, Marques MM, Beland FA. Induction of lacI mutations in Big Blue rats treated with tamoxifen and alpha-hydroxytamoxifen. Cancer Lett. 2002;176:37–45. [PubMed: 11790452] [CrossRef]
- Gamboa da Costa G, Marques MM, Freeman JP, Beland FA. Synthesis and investigation of alpha-hydroxy-N,N-didesmethyltamoxifen as a proximate carcinogen in the metabolic activation of tamoxifen. Chem Res Toxicol. 2003;16:1090–1098. [PubMed: 12971796] [CrossRef]
- Gamboa da Costa G, McDaniel-Hamilton LP, Heflich RH, et al. DNA adduct formation and mutant induction in Sprague-Dawley rats treated with tamoxifen and its derivatives. Carcinogenesis. 2001;22:1307–1315. [PubMed: 11470763] [CrossRef]
- Gamboa da Costa G, Pereira PC, Churchwell MI, et al. DNA adduct formation in the livers of female Sprague-Dawley rats treated with toremifene or α-hydroxytoremifene. Chem Res Toxicol. 2007;20:300–310. [PubMed: 17261033] [CrossRef]
- Glatt H, Bartsch I, Christoph S, et al. Sulfotransferase-mediated activation of mutagens studied using heterologous expression systems. Chem Biol Interact. 1998;109:195–219. b. [PubMed: 9566746] [CrossRef]
- Glatt H, Davis W, Meinl W, et al. Rat, but not human, sulfotransferase activates a tamoxifen metabolite to produce DNA adducts and gene mutations in bacteria and mammalian cells in culture. Carcinogenesis. 1998;19:1709–1713. a. [PubMed: 9806149] [CrossRef]
- Greaves P, Goonetilleke R, Nunn G, et al. Two-year carcinogenicity study of tamoxifen in Alderley Park Wistar-derived rats. Cancer Res. 1993;53:3919–3924. [PubMed: 8358718]
- Green AR, Styles JA, Parrott EL, et al. Neonatal tamoxifen treatment of mice leads to adenomyosis but not uterine cancer. Exp Toxicol Pathol. 2005;56:255–263. [PubMed: 15816354] [CrossRef]
- Hachisuga T, Tsujioka H, Horiuchi S, et al. K-ras mutation in the endometrium of tamoxifen-treated breast cancer patients, with a comparison of tamoxifen and toremifene. Br J Cancer. 2005;92:1098–1103. [PMC free article: PMC2361944] [PubMed: 15756272] [CrossRef]
- Halakivi-Clarke L, Cho E, Onojafe I, et al. Maternal exposure to tamoxifen during pregnancy increases carcinogen-induced mammary tumorigenesis among female rat offspring. Clin Cancer Res. 2000;6:305–308. [PubMed: 10656462]
- Hard GC, Iatropoulos MJ, Jordan K, et al. Major difference in the hepatocarcinogenicity and DNA adduct forming ability between toremifene and tamoxifen in female Crl:CD(BR) rats. Cancer Res. 1993;53:4534–4541. [PubMed: 8402624]
- Hardcastle IR, Horton MN, Osborne MR, et al. Synthesis and DNA reactivity of alpha-hydroxylated metabolites of nonsteroidal antiestrogens. Chem Res Toxicol. 1998;11:369–374. [PubMed: 9548808] [CrossRef]
- Hasmann M, Rattel B, Löser R. Preclinical data for Droloxifene. Cancer Lett. 1994;84:101–116. [PubMed: 8076367] [CrossRef]
- Hemminki K, Rajaniemi H, Koskinen M, Hansson J. Tamoxifen-induced DNA adducts in leucocytes of breast cancer patients. Carcinogenesis. 1997;18:9–13. [PubMed: 9054583] [CrossRef]
- Hemminki K, Rajaniemi H, Lindahl B, Moberger B. Tamoxifen-induced DNA adducts in endometrial samples from breast cancer patients. Cancer Res. 1996;56:4374–4377. [PubMed: 8813128]
- Hirsimäki P, Hirsimäki Y, Nieminen L, Payne BJ. Tamoxifen induces hepatocellular carcinoma in rat liver: a 1-year study with two antiestrogens. Arch Toxicol. 1993;67:49–54. [PubMed: 8452480] [CrossRef]
- IARC. Some pharmaceutical drugs. IARC Monogr Eval Carcinog Risks Hum. 1996;66:1–514.
- Jones LP, Li M, Halama ED, et al. Promotion of mammary cancer development by tamoxifen in a mouse model of Brca1-mutation-related breast cancer. Oncogene. 2005;24:3554–3562. [PubMed: 15750629] [CrossRef]
- Jordan VC, Lababidi MK, Langan-Fahey S. Suppression of mouse mammary tumorigenesis by long-term tamoxifen therapy. J Natl Cancer Inst. 1991;83:492–496. [PubMed: 2005632] [CrossRef]
- Jordan VC, Lababidi MK, Mirecki DM. Anti-oestrogenic and anti-tumour properties of prolonged tamoxifen therapy in C3H/OUJ mice. Eur J Cancer. 1990;26:718–721. [PubMed: 2144160] [CrossRef]
- Karim BO, Landolfi JA, Christian A, et al. Estrous cycle and ovarian changes in a rat mammary carcinogenesis model after irradiation, tamoxifen chemoprevention, and aging. Comp Med. 2003;53:532–538. [PubMed: 14655997]
- Kärki A, Mäntylä E, Hirsimäki Y, et al. Comparison of the effects of tamoxifen and toremifene on rat hepatocarcinogenesis. Arch Toxicol. 2000;74:249–256. [PubMed: 10959800] [CrossRef]
- Karlsson S. Histopathology and histomorphometry of the urogenital tract in 15-month old male and female rats treated neonatally with SERMs and estrogens. Exp Toxicol Pathol. 2006;58:1–12. [PubMed: 16709447] [CrossRef]
- Kasahara T, Kuwayama C, Hashiba M, et al. The gene expression of hepatic proteins responsible for DNA repair and cell proliferation in tamoxifen-induced hepatocarcinogenesis. Cancer Sci. 2003;94:582–588. [PubMed: 12841865] [CrossRef]
- Katase K, Sugiyama Y, Hasumi K, et al. The incidence of subsequent endometrial carcinoma with tamoxifen use in patients with primary breast carcinoma. Cancer. 1998;82:1698–1703. [PubMed: 9576291] [CrossRef]
- Kim SY, Laxmi YR, Suzuki N, et al. Formation of tamoxifen-DNA adducts via O-sulfonation, not O-acetylation, of alpha-hydroxytamoxifen in rat and human livers. Drug Metab Dispos. 2005;33:1673–1678. [PubMed: 16099924] [CrossRef]
- Kim SY, Suzuki N, Laxmi YR, et al. Antiestrogens and the formation of DNA damage in rats: a comparison. Chem Res Toxicol. 2006;19:852–858. a. [PMC free article: PMC2533849] [PubMed: 16780365] [CrossRef]
- Kim SY, Suzuki N, Laxmi YR, Shibutani S. Inefficient repair of tamoxifen-DNA adducts in rats and mice. Drug Metab Dispos. 2006;34:311–317. b. [PubMed: 16299164] [CrossRef]
- Kim SY, Suzuki N, Santosh Laxmi YR, et al. Alpha-hydroxylation of tamoxifen and toremifene by human and rat cytochrome P450 3A subfamily enzymes. Chem Res Toxicol. 2003;16:1138–1144. [PubMed: 12971802] [CrossRef]
- Kitagawa M, Ravindernath A, Suzuki N, et al. Identification of tamoxifen-DNA adducts induced by alpha-acetoxy-N-desmethyltamoxifen. Chem Res Toxicol. 2000;13:761–769. [PubMed: 10956064] [CrossRef]
- Lavie O, Barnett-Griness O, Narod SA, Rennert G. The risk of developing uterine sarcoma after tamoxifen use. Int J Gynecol Cancer. 2008;18:352–356. [PubMed: 18334013] [CrossRef]
- Lecce G, Meduri G, Ancelin M, et al. Presence of estrogen receptor β in the human endometrium through the cycle: expression in glandular, stromal, and vascular cells. J Clin Endocrinol Metab. 2001;86:1379–1386. [PubMed: 11238535] [CrossRef]
- Li D, Dragan Y, Jordan VC, et al. Effects of chronic administration of tamoxifen and toremifene on DNA adducts in rat liver, kidney, and uterus. Cancer Res. 1997;57:1438–1441. [PubMed: 9108442]
- Lien EA, Solheim E, Lea OA, et al. Distribution of 4-hydroxy-N-desmethyltamoxifen and other tamoxifen metabolites in human biological fluids during tamoxifen treatment. Cancer Res. 1989;49:2175–2183. [PubMed: 2702659]
- Lien EA, Solheim E, Ueland PM. Distribution of tamoxifen and its metabolites in rat and human tissues during steady-state treatment. Cancer Res. 1991;51:4837–4844. [PubMed: 1893376]
- Maltoni C, Minardi F, Pinto C, et al. Results of three life-span experimental carcinogenicity and anticarcinogenicity studies on tamoxifen in rats. Ann N Y Acad Sci. 1997;837:469–512. [PubMed: 9472359] [CrossRef]
- Mani C, Gelboin HV, Park SS, et al. Metabolism of the antimammary cancer antiestrogenic agent tamoxifen. I. Cytochrome P-450-catalyzed N-demethylation and 4-hydroxylation. Drug Metab Dispos. 1993;21:645–656. [PubMed: 8104124]
- Mani C, Kupfer D. Cytochrome P-450-mediated activation and irreversible binding of the antiestrogen tamoxifen to proteins in rat and human liver: possible involvement of flavin-containing monooxygenases in tamoxifen activation. Cancer Res. 1991;51:6052–6058. [PubMed: 1933868]
- Mani C, Pearce R, Parkinson A, Kupfer D. Involvement of cytochrome P4503A in catalysis of tamoxifen activation and covalent binding to rat and human liver microsomes. Carcinogenesis. 1994;15:2715–2720. [PubMed: 8001226] [CrossRef]
- Mäntylä ETE, Karlsson SH, Nieminen LS (1996). Induction of Endometrial Cancer by Tamoxifen in the rat. In: Hormonal Carcinogenesis II Proceedings of the 2nd International Symposium on Hormonal Carcinogenesis. Li JJ, Li SA, Gustafsson JA et al., editors. New York: Springer Verlag, pp. 442–445.
- Marques MM, Beland FA. Identification of tamoxifen-DNA adducts formed by 4-hydroxytamoxifen quinone methide. Carcinogenesis. 1997;18:1949–1954. [PubMed: 9364005] [CrossRef]
- Martin EA, Brown K, Gaskell M, et al. Tamoxifen DNA damage detected in human endometrium using accelerator mass spectrometry. Cancer Res. 2003;63:8461–8465. [PubMed: 14679010]
- Martin EA, Carthew P, White IN, et al. Investigation of the formation and accumulation of liver DNA adducts in mice chronically exposed to tamoxifen. Carcinogenesis. 1997;18:2209–2215. [PubMed: 9395223] [CrossRef]
- Martin EA, Heydon RT, Brown K, et al. Evaluation of tamoxifen and alpha-hydroxytamoxifen 32P-post-labelled DNA adducts by the development of a novel automated on-line solid-phase extraction HPLC method. Carcinogenesis. 1998;19:1061–1069. [PubMed: 9667745] [CrossRef]
- Martin EA, Rich KJ, White IN, et al. 32P-postlabelled DNA adducts in liver obtained from women treated with tamoxifen. Carcinogenesis. 1995;16:1651–1654. [PubMed: 7614701] [CrossRef]
- Matsuyama Y, Tominaga T, Nomura Y, et al. Second cancers after adjuvant tamoxifen therapy for breast cancer in Japan. Ann Oncol. 2000;11:1537–1543. [PubMed: 11205460] [CrossRef]
- McLuckie KI, Crookston RJ, Gaskell M, et al. Mutation spectra induced by alpha-acetoxytamoxifen-DNA adducts in human DNA repair proficient and deficient (xeroderma pigmentosum complementation group A) cells. Biochemistry. 2005;44:8198–8205. [PubMed: 15924439] [CrossRef]
- McLuckie KI, Routledge MN, Brown K, et al. DNA adducts formed from 4-hydroxytamoxifen are more mutagenic than those formed by alpha-acetoxytamoxifen in a shuttle vector target gene replicated in human Ad293 cells. Biochemistry. 2002;41:8899–8906. [PubMed: 12102632] [CrossRef]
- Metcalfe KA, Lynch HT, Ghadirian P, et al. The risk of ovarian cancer after breast cancer in BRCA1 and BRCA2 carriers. Gynecol Oncol. 2005;96:222–226. [PubMed: 15589605] [CrossRef]
- Mignotte H, Lasset C, Bonadona V, et al. Fédération Nationale des Centres de Lutte Contre le Cancer (FNCLCC). Iatrogenic risks of endometrial carcinoma after treatment for breast cancer in a large French case-control study. Int J Cancer. 1998;76:325–330. [PubMed: 9579567] [CrossRef]
- Mourits MJ, Hollema H, De Vries EG, et al. Apoptosis and apoptosis-associated parameters in relation to tamoxifen exposure in postmenopausal endometrium. Hum Pathol. 2002;33:341–346. [PubMed: 11979376] [CrossRef]
- Newbold RR, Jefferson WN, Padilla-Burgos E, Bullock BC. Uterine carcinoma in mice treated neonatally with tamoxifen. Carcinogenesis. 1997;18:2293–2298. [PubMed: 9450472] [CrossRef]
- Newcomb PA, Solomon C, White E. Tamoxifen and risk of large bowel cancer in women with breast cancer. Breast Cancer Res Treat. 1999;53:271–277. [PubMed: 10369073] [CrossRef]
- Notley LM, Crewe KH, Taylor PJ, et al. Characterization of the human cytochrome P450 forms involved in metabolism of tamoxifen to its alpha-hydroxy and alpha,4-dihydroxy derivatives. Chem Res Toxicol. 2005;18:1611–1618. [PubMed: 16533026] [CrossRef]
- O’Neil MJ, editor (2006). The Merck Index, 14th ed. Whitehouse Station, NJ: Merck & Co., Inc., p. 1554.
- Osborne MR, Davis W, Hewer AJ, et al. 4-Hydroxytamoxifen gives DNA adducts by chemical activation, but not in rat liver cells. Chem Res Toxicol. 1999;12:151–158. [PubMed: 10027792] [CrossRef]
- Osborne MR, Hardcastle IR, Phillips DH. Minor products of reaction of DNA with alpha-acetoxytamoxifen. Carcinogenesis. 1997;18:539–543. [PubMed: 9067554] [CrossRef]
- Osborne MR, Hewer A, Hardcastle IR, et al. Identification of the major tamoxifen-deoxyguanosine adduct formed in the liver DNA of rats treated with tamoxifen. Cancer Res. 1996;56:66–71. [PubMed: 8548777]
- Osborne MR, Hewer A, Phillips DH. Resolution of alpha-hydroxytamoxifen; R-isomer forms more DNA adducts in rat liver cells. Chem Res Toxicol. 2001;14:888–893. [PubMed: 11453736] [CrossRef]
- Osborne MR, Hewer A, Phillips DH. Stereoselective metabolic activation of alpha-hydroxy-N-desmethyltamoxifen: the R-isomer forms more DNA adducts in rat liver cells. Chem Res Toxicol. 2004;17:697–701. [PubMed: 15144227] [CrossRef]
- Pathak DN, Pongracz K, Bodell WJ. Microsomal and peroxidase activation of 4-hydroxy-tamoxifen to form DNA adducts: comparison with DNA adducts formed in Sprague-Dawley rats treated with tamoxifen. Carcinogenesis. 1995;16:11–15. [PubMed: 7834794] [CrossRef]
- Pathak DN, Pongracz K, Bodell WJ. Activation of 4-hydroxytamoxifen and the tamoxifen derivative metabolite E by uterine peroxidase to form DNA adducts: comparison with DNA adducts formed in the uterus of Sprague-Dawley rats treated with tamoxifen. Carcinogenesis. 1996;17:1785–1790. [PubMed: 8824496] [CrossRef]
- Phillips DH, Carmichael PL, Hewer A, et al. Activation of tamoxifen and its metabolite alpha-hydroxytamoxifen to DNA-binding products: comparisons between human, rat and mouse hepatocytes. Carcinogenesis. 1996;17:89–94. a. [PubMed: 8565142] [CrossRef]
- Phillips DH, Hewer A, Grover PL, et al. Tamoxifen does not form detectable DNA adducts in white blood cells of breast cancer patients. Carcinogenesis. 1996;17:1149–1152. b. [PubMed: 8640926] [CrossRef]
- Phillips DH, Hewer A, Horton MN, et al. N-demethylation accompanies alpha-hydroxylation in the metabolic activation of tamoxifen in rat liver cells. Carcinogenesis. 1999;20:2003–2009. [PubMed: 10506117] [CrossRef]
- Phillips DH, Hewer A, Osborne MR, et al. Organ specificity of DNA adduct formation by tamoxifen and alpha-hydroxytamoxifen in the rat: implications for understanding the mechanism(s) of tamoxifen carcinogenicity and for human risk assessment. Mutagenesis. 2005;20:297–303. [PubMed: 15928012] [CrossRef]
- Pole J, Carmichael P, Griffin J. Identification of transcriptional biomarkers induced by SERMS in human endometrial cells using multivariate analysis of DNA microarrays. Biomarkers. 2004;9:447–460. [PubMed: 15849065] [CrossRef]
- Pole JC, Gold LI, Orton T, et al. Gene expression changes induced by estrogen and selective estrogen receptor modulators in primary-cultured human endometrial cells: signals that distinguish the human carcinogen tamoxifen. Toxicology. 2005;206:91–109. [PubMed: 15590111] [CrossRef]
- Poon GK, Chui YC, McCague R, et al. Analysis of phase I and phase II metabolites of tamoxifen in breast cancer patients. Drug Metab Dispos. 1993;21:1119–1124. [PubMed: 7905393]
- Poon GK, Walter B, Lønning PE, et al. Identification of tamoxifen metabolites in human Hep G2 cell line, human liver homogenate, and patients on long-term therapy for breast cancer. Drug Metab Dispos. 1995;23:377–382. [PubMed: 7628304]
- Rajaniemi H, Koskinen M, Mäntylä E, Hemminki K. DNA binding of tamoxifen and its analogues: identification of the tamoxifen-DNA adducts in rat liver. Toxicol Lett. 1998;102-103:453–457. [PubMed: 10022295] [CrossRef]
- Rajaniemi H, Rasanen I, Koivisto P, et al. Identification of the major tamoxifen-DNA adducts in rat liver by mass spectroscopy. Carcinogenesis. 1999;20:305–309. [PubMed: 10069469] [CrossRef]
- Razvi N, Greaves P, Styles J, et al. Absence of uterine tumours in CD-1 mice treated neonatally with subcutaneous tamoxifen or 4-hydroxyoestradiol. Exp Toxicol Pathol. 2007;59:177–185. [PubMed: 17825543] [CrossRef]
- Royal Pharmaceutical Society of Great Britain (2007). British National Formulary, No. 54. London: BMJ Publishing Group Ltd./RPS Publishing.
- Rutqvist LE, Johansson H., Stockholm Breast Cancer Study Group. Long-term follow-up of the randomized Stockholm trial on adjuvant tamoxifen among postmenopausal patients with early stage breast cancer. Acta Oncol. 2007;46:133–145. [PubMed: 17453361] [CrossRef]
- Sauvez F, Drouin DS, Attia M, et al. Cutaneously applied 4-hydroxytamoxifen is not carcinogenic in female rats. Carcinogenesis. 1999;20:843–850. [PubMed: 10334202] [CrossRef]
- Schild LJ, Divi RL, Beland FA, et al. Formation of tamoxifen-DNA adducts in multiple organs of adult female cynomolgus monkeys dosed with tamoxifen for 30 days. Cancer Res. 2003;63:5999–6003. [PubMed: 14522927]
- Shang Y. Molecular mechanisms of oestrogen and SERMs in endometrial carcinogenesis. Nat Rev Cancer. 2006;6:360–368. [PubMed: 16633364] [CrossRef]
- Sharma M, Shubert DE, Sharma M, et al. Antioxidant inhibits tamoxifen-DNA adducts in endometrial explant culture. Biochem Biophys Res Commun. 2003;307:157–164. [PubMed: 12849995] [CrossRef]
- Sherman ME. Theories of endometrial carcinogenesis: a multidisciplinary approach. Mod Pathol. 2000;13:295–308. [PubMed: 10757340] [CrossRef]
- Shibutani S, Dasaradhi L. Miscoding potential of tamoxifen-derived DNA adducts: alpha-(N2-deoxyguanosinyl)tamoxifen. Biochemistry. 1997;36:13010–13017. [PubMed: 9335562] [CrossRef]
- Shibutani S, Dasaradhi L, Terashima I, et al. Alpha-hydroxytamoxifen is a substrate of hydroxysteroid (alcohol) sulfotransferase, resulting in tamoxifen DNA adducts. Cancer Res. 1998;58:647–653. b. [PubMed: 9485016]
- Shibutani S, Ravindernath A, Suzuki N, et al. Identification of tamoxifen-DNA adducts in the endometrium of women treated with tamoxifen. Carcinogenesis. 2000;21:1461–1467. a. [PubMed: 10910945] [CrossRef]
- Shibutani S, Reardon JT, Suzuki N, Sancar A. Excision of tamoxifen-DNA adducts by the human nucleotide excision repair system. Cancer Res. 2000;60:2607–2610. b. [PubMed: 10825130]
- Shibutani S, Shaw PM, Suzuki N, et al. Sulfation of alpha-hydroxytamoxifen catalyzed by human hydroxysteroid sulfotransferase results in tamoxifen-DNA adducts. Carcinogenesis. 1998;19:2007–2011. a. [PubMed: 9855017] [CrossRef]
- Shibutani S, Suzuki N, Laxmi YR, et al. Identification of tamoxifen-DNA adducts in monkeys treated with tamoxifen. Cancer Res. 2003;63:4402–4406. [PubMed: 12907611]
- Shibutani S, Suzuki N, Terashima I, et al. Tamoxifen-DNA adducts detected in the endometrium of women treated with tamoxifen. Chem Res Toxicol. 1999;12:646–653. [PubMed: 10409405] [CrossRef]
- Singh MN, Stringfellow HF, Walsh MJ, et al. Quantifiable mRNA transcripts for tamoxifen-metabolising enzymes in human endometrium. Toxicology. 2008;249:85–90. [PubMed: 18502016] [CrossRef]
- Srinivasan R, Yang YX, Rubin SC, et al. Women with a prior diagnosis of breast cancer are not at an increased risk for subsequent colorectal cancer. Am J Gastroenterol. 2005;100:2759–2764. [PubMed: 16393232] [CrossRef]
- Styles JA, Davies A, Davies R, et al. Clastogenic and aneugenic effects of tamoxifen and some of its analogues in hepatocytes from dosed rats and in human lymphoblastoid cells transfected with human P450 cDNAs (MCL-5 cells). Carcinogenesis. 1997;18:303–313. [PubMed: 9054622] [CrossRef]
- Styles JA, Davies R, Fenwick S, et al. Tamoxifen mutagenesis and carcinogenesis in livers of lambda/lacI transgenic rats: selective influence of phenobarbital promotion. Cancer Lett. 2001;162:117–122. [PubMed: 11121869] [CrossRef]
- Swerdlow AJ, Jones ME. Ovarian cancer risk in premenopausal and perimenopausal women treated with Tamoxifen: a case-control study. Br J Cancer. 2007;96:850–855. [PMC free article: PMC2360088] [PubMed: 17285129] [CrossRef]
- Swerdlow AJ, Jones ME., British Tamoxifen Second Cancer Study Group. Tamoxifen treatment for breast cancer and risk of endometrial cancer: a case-control study. J Natl Cancer Inst. 2005;97:375–384. [PubMed: 15741574] [CrossRef]
- Terashima I, Suzuki N, Shibutani S. Mutagenic potential of alpha-(N2-deoxyguanosinyl)tamoxifen lesions, the major DNA adducts detected in endometrial tissues of patients treated with tamoxifen. Cancer Res. 1999;59:2091–2095. [PubMed: 10232593]
- The Early Breast Cancer Trialists’ Collaborative Group. Tamoxifen for early breast cancer: an overview of the randomised trials. Lancet. 1998;351:1451–1467. [PubMed: 9605801] [CrossRef]
- Thomson Healthcare (2007). Physicians’ Desk Reference, 61st ed. Montvale, NJ: Thomson, pp. 2149, 3527–3533.
- Tucker MJ, Adam HK, Patterson JS (1984). Tamoxifen. In: Safety Testing of New Drugs. Laurence DR, McLean AEM, Weatherall M, editors. New York: Academic Press, pp. 125–162.
- Umemoto A, Komaki K, Monden Y, et al. Identification and quantification of tamoxifen-DNA adducts in the liver of rats and mice. Chem Res Toxicol. 2001;14:1006–1013. [PubMed: 11511174] [CrossRef]
- Umemoto A, Monden Y, Komaki K, et al. Tamoxifen-DNA adducts formed by alpha-acetoxytamoxifen N-oxide. Chem Res Toxicol. 1999;12:1083–1089. [PubMed: 10563834] [CrossRef]
- Umemoto A, Monden Y, Lin CX, et al. Determination of tamoxifen–DNA adducts in leukocytes from breast cancer patients treated with tamoxifen. Chem Res Toxicol. 2004;17:1577–1583. [PubMed: 15606132] [CrossRef]
- Umemoto A, Monden Y, Suwa M, et al. Identification of hepatic tamoxifen-DNA adducts in mice: alpha-(N(2)-deoxyguanosinyl)tamoxifen and alpha-(N(2)-deoxyguanosinyl)tamoxifen N-oxide. Carcinogenesis. 2000;21:1737–1744. [PubMed: 10964106] [CrossRef]
- Ursic Vrscaj M, Kovacic J, Bebar S, et al. Endometrial and other primary cancers after tamoxifen treatment of breast cancer – results of retrospective cohort study. Eur J Obstet Gynecol Reprod Biol. 2001;95:105–110. [PubMed: 11267730] [CrossRef]
- Waalkes MP, Liu J, Ward JM, et al. Urogenital carcinogenesis in female CD1 mice induced by in utero arsenic exposure is exacerbated by postnatal diethylstilbestrol treatment. Cancer Res. 2006;66:1337–1345. a. [PubMed: 16452187] [CrossRef]
- Waalkes MP, Liu J, Ward JM, Diwan BA. Enhanced urinary bladder and liver carcinogenesis in male CD1 mice exposed to transplacental inorganic arsenic and postnatal diethylstilbestrol or tamoxifen. Toxicol Appl Pharmacol. 2006;215:295–305. b. [PubMed: 16712894] [CrossRef]
- Wallén M, Tomás E, Visakorpi T, et al. Endometrial K-ras mutations in postmenopausal breast cancer patients treated with adjuvant tamoxifen or toremifene. Cancer Chemother Pharmacol. 2005;55:343–346. [PubMed: 15592834] [CrossRef]
- White IN, Carthew P, Davies R, et al. Short-term dosing of alpha-hydroxytamoxifen results in DNA damage but does not lead to liver tumours in female Wistar/Han rats. Carcinogenesis. 2001;22:553–557. [PubMed: 11285188] [CrossRef]
- White IN, Martin EA, Mauthe RJ, et al. Comparisons of the binding of [14C]radiolabelled tamoxifen or toremifene to rat DNA using accelerator mass spectrometry. Chem Biol Interact. 1997;106:149–160. [PubMed: 9366900] [CrossRef]
- Williams GM, Iatropoulos MJ, Djordjevic MV, Kaltenberg OP. The triphenylethylene drug tamoxifen is a strong liver carcinogen in the rat. Carcinogenesis. 1993;14:315–317. [PubMed: 8435874] [CrossRef]
- Williams GM, Iatropoulos MJ, Karlsson S. Initiating activity of the anti-estrogen tamoxifen, but not toremifene in rat liver. Carcinogenesis. 1997;18:2247–2253. [PubMed: 9395228] [CrossRef]
- Wolff AC, Abeloff MD. Adjuvant chemotherapy for postmenopausal lymph node-negative breast cancer: it ain’t necessarily so. J Natl Cancer Inst. 2002;94:1041–1043. [PubMed: 12122089]
- Wozniak K, Kolacinska A, Blasinska-Morawiec M, et al. The DNA-damaging potential of tamoxifen in breast cancer and normal cells. Arch Toxicol. 2007;81:519–527. [PubMed: 17593413] [CrossRef]
- Wu H, Chen Y, Liang J, et al. Hypomethylation-linked activation of PAX2 mediates tamoxifen-stimulated endometrial carcinogenesis. Nature. 2005;438:981–987. [PubMed: 16355216] [CrossRef]
- Yadollahi-Farsani M, Davies DS, Boobis AR. The mutational signature of alpha-hydroxytamoxifen at Hprt locus in Chinese hamster cells. Carcinogenesis. 2002;23:1947–1952. [PubMed: 12419845] [CrossRef]
- Yamazawa K, Miyazawa Y, Suzuki M, et al. Tamoxifen and the risk of endometrial cancer in Japanese women with breast cancer. Surg Today. 2006;36:41–46. [PubMed: 16378192] [CrossRef]
- Yasui M, Suzuki N, Laxmi YR, Shibutani S. Translesion synthesis past tamoxifen-derived DNA adducts by human DNA polymerases eta and kappa. Biochemistry. 2006;45:12167–12174. [PMC free article: PMC2593916] [PubMed: 17002316] [CrossRef]
- TAMOXIFEN - PharmaceuticalsTAMOXIFEN - Pharmaceuticals
Your browsing activity is empty.
Activity recording is turned off.
See more...