Download Major Difference in the Hepatocarcinogenicity

Survey
yes no Was this document useful for you?
   Thank you for your participation!

* Your assessment is very important for improving the workof artificial intelligence, which forms the content of this project

Document related concepts
no text concepts found
Transcript
[CANCER RESEARCH 53, 4534--4541, October l, 1993]
Major Difference in the Hepatocarcinogenicity and DNA Adduct Forming Ability
between Toremifene and Tamoxifen in Female CrhCD(BR) Rats 1
G o r d o n C. H a r d , 2 M i c h a e l J. I a t r o p o u l o s , K e v i n J o r d a n , L e i l a R a d i , O l g i e r d P. K a l t e n b e r g ,
Anthony R. Imondi, and Gary M. Williams
American Health Foundation, Valhalla, New York 10595 [G. (7. H., M. J. L, K. J., L. R., 0 P. K., G. M. W.], and Adria Laboratories, Columbus, Ohio 43216 [A. R. L]
ABSTRACT
The hepatoproliferative effects of 2 antiestrogens, tamoxifen and toremifene, were compared in a sequential 15-month study in which 2 doses of
each compound were administered by daily gavage to female SpragueDawley rats for up to 12 months. The doses were 11.3 and 22.6 mg/kg for
tamoxifen and 12 and 24 mg/kg for toremifene. There were scheduled
sacrifices at 3, 6, 12, and 15 months, the latter including a 3-month
recovery period from the 12th through the 14th month. In the chronic
toxicity study, tamoxifen at 22.6 mg/kg produced 100% incidence of hepatoceUular carcinoma at the 12- and 15-month sacrifice intervals and
67% and 71% incidences at the ll.3.mg/kg dose. Sequential observations
showed an increased incidence of glutathione S-transferase-positive foci of
hepatocellular alteration by 3 months with tamoxifen in the absence of
hepatotoxicity, with the first liver carcinoma appearing by 6 months of
treatment. Unscheduled deaths occurring beyond 7.5 months in the
tamoxifen treated groups were due in almost all cases to liver cancer. In
striking contrast, toremifene did not produce any hepatoproliferative effects at 12- and 24-mg/kg dose levels, nor in a pilot study at 48 mg/kg. The
24.mg/kg dose of toremifene exerted an inhibiting effect on loci of hepatocellular alteration in rat liver detectable by glutathione S-transferase
immunohistochemistry at 3 months and by conventional histology at 12
months. An antiproliferative effect was also evident in mammary gland
and anterior pituitary where both toremifene and tamoxifen suppressed
tumor incidence in comparison to the control group. The ability of these
drugs to modify rat liver DNA after p.o. administration was investigated
using the a2p-postlabeling assay. Administration of tamoxifen at 45 mg/kg
for 7 days produced liver DNA nucleoside modifications represented by 7
spots on the autoradiogram. Unlike tamoxifen, toremifene did not produce
any modified bases in rat liver DNA detectable by the 32p-postlabeling
technique. The dose levels of tamoxifen that are strongly hepatocarcinogenic in the rat are compared with doses used in humans in various
applications. Taking internal drug exposure into account, we
conclude that the margin of safety for use of tamoxifen as an endocrine prophylactic agent for healthy, but breast cancer prone, women is
questionable.
INTRODUCTION
Tamoxifen and toremifene are nonsteroidal triphenylethylene derivatives with chemical structures closely related to diethylstilbestrol
(Fig. 1). They both have antiestrogenic and estrogenic properties (1,
2), depending on species and tissue type. Tamoxifen has been used
extensively in the treatment of breast cancer, both in advanced cases
and more recently as adjuvant treatment in early-stage disease (3, 4).
Because women treated with tamoxifen were found to have a lower
incidence of second and contralateral breast cancer, the possibility that
tamoxifen could be an effective endocrine cancer preventive agent in
women at increased risk of breast cancer has been advocated (5-9).
Consequently, cancer prophylaxis trials in the United States, United
Kingdom, and Italy have been initiated on groups of women at high
Received4/8/93; accepted7/26/93.
The costs of publicationof this article were defrayedin part by the paymentof page
charges. This article must thereforebe hereby markedadvertisement in accordancewith
18 U.S.C. Section 1734 solely to indicatethis fact.
1This researchwas supportedfinanciallyby Adria Laboratories, Columbus, OH, and
Farmos Group, Ltd., Turku, Finland.
z To whomrequestsfor reprints shouldbe addressed,at AmericanHealthFoundation,
1 Dana Road, Valhalla, NY 10595.
risk for developing breast cancer (8, 10, 11). This trial application as
a chemopreventive in healthy women has occurred despite the knowledge that tamoxifen produced liver tumors in rats in the 2-year safety
evaluation study conducted by the manufacturer (12).
Toremifene has also been developed as a breast cancer treatment
(13). It differs from tamoxifen only by the substitution of a chlorine
atom for a hydrogen atom in an ethyl chain attached to one of the
carbon atoms of the ethylene bond (Fig. 1). Toremifene has antiestrogenic and estrogenic properties similar to those of tamoxifen (1, 2,
14, 15), although it is cytostatic for human breast cancer cells at a dose
that is 3 times higher than for tamoxifen (15). Currently toremifene
is undergoing clinical evaluation in Europe and the United States
(15-18).
Recently tamoxifen was shown to be strongly hepatocarcinogenic
to female rats (19, 20), confirming an observation noted in the manufacturer's safety evaluation report (12). Also, tamoxifen has been
reported to be DNA-damaging (21, 22). The present nonclinical study
was undertaken to compare the chronic effects of tamoxifen and
toremifene under conditions of GLP, 3 specifically to determine
whether toremifene shared the potent hepatocarcinogenic properties of
its analogue. The chronic toxicity study design was a 1-year daily
gavage trial with interim sacrifices, and including a 3-month recovery
period. In addition, DNA adduct formation was evaluated in rat liver
exposed in vivo to both compounds for 7 consecutive days using the
32p-postlabeling technique to measure modified bases (23).
M A T E R I A L S AND M E T H O D S
Chemicals. Tamoxifen citrate and toremifene citrate, each of 99% purity,
were obtained from Adria Laboratories, Columbus, OH, and protected from
light during storage. Fresh suspensions of the test compounds in 0.5% carboxymethylcellulose(Sigma, St. Louis, MO) were prepared each week and the
concentration checked by UV-spectrophotometry on a monthly basis. Each
compound was administered at 2 dose levels: 11.3 mg/kg (concentration 2.3
mg/ml) and 22.6 mg/kg (4.5 mg/ml) for tamoxifen, and 12 mg/kg (2.4 mg/ml)
and 24 mg/kg (4.8 mg/ml) for toremifene. The dosing volume for each drug
suspension was 5 ml/kg body weight.
Test Animals and Animal Care. Female CrI:CD(BR) rats (Sprague-Dawley strain) aged approximately 4 weeks were obtained from Charles River
Laboratories, Kingston, NY, for the carcinogenicity study. After acclimation
and observation for 2 weeks, the animals were individually identified and
assigned to groups using a randomization procedure based on body weights.
The animals were identified by ear notch and toe clip and were housed 3 per
cage on standard hardwood commercial rodent bedding (Beta Chip; Northeastern Products Corp., Warrensburg, NY) in temperature- (71 _ 5~ and
humidity- (55 +-- 15%) controlled rooms with 12-h dark/low-level fluorescent
light cycles changing at 7 a.m. and 7 p.m. Cage rack rotation was performed
every 2 weeks. The animals were provided with NIH-07 pelleted diet (Purina
Mills, Inc., St. Louis, MO) and water ad libitum supplied by Westchester
County through an automated watering system. Certificates of analysis for diet,
drinking water, and bedding, covering a standard range of microbial contaminants, inorganic elements, chlorinated hydrocarbons, organophosphates,
3 The abbreviations used are: GLP, good laboratory practice; H&E, hematoxylin and
eosin; GST-P, glutathione S-transferase, placental form; DMBA, 7,12-dimethylbenzanthracene.
4534
Downloaded from cancerres.aacrjournals.org on June 15, 2017. © 1993 American Association for Cancer
Research.
LIVER EFFECTS OF TOREMIFENE AND TAMOXIFEN
caac~a2 /~rOH
0H~
C=C "~-'~
\CH2CH3
Diethylstilbestrol
CH3"NCH CHoO-Z~
CH3 /
2
~
/=X
~',C:Ct~k ~,
\cHzCH3
Tamoxifen
CH3 ~,NCH2CH20
CH3/
"~C=CO
CH2CH2CI
Toremifene
Fig. 1. Chemical structures of diethylstilbestrol, tamoxifen, and toremifene.
nitrosamines, and aflatoxins, indicated compliance within recommended contaminant limits (24, 25).
All animal maintenance and handling procedures were conducted in accordance with United States Department of Agriculture and NIH guidelines for
humane care. The animal protocol was approved by the Institutional Animal
Care and Use Committee prior to commencement. The American Health Foundation research animal facility is fully accredited by the American Association
for the Accreditation of Laboratory Animal Care.
Carcinogenicity Study Design. A total of 375 animals were assigned to
one control group and 2 dose-level groups for each test compound as summarized in Table 1. Three animals from each treatment group served as sentinels
for health monitoring. There were 4 scheduled sacrifice intervals: 3 interim at
3, 6, and 12 months, and a terminal sacrifice at 15 months.
The animals were dosed p.o. by gavage once daily, 7 days a week for up to
12 months. Animals assigned to the 15-month sacrifice groups received no
further treatment beyond 12 months, the final 3 months representing a recovery
period.
The results from a pilot study on toremifene are also included in this report
because the data extend the upper dose-range of toremifene tested. In the pilot
study, a 48-mg/kg dose of toremifene citrate was administered by daily gavage
to female Sprague-Dawley rats for 12 months before termination. There was an
interim sacrifice at 6-7 months. The conditions of this pilot study were identical to the bioassay described above except that it was not audited according
to GLP. The numbers of animals are shown in Table l.
Clinical Observations. Clinical observations for toxic or pharmacological
effects were conducted daily and body weights recorded weekly for 8 weeks,
biweekly for the next 16 weeks, and monthly thereafter for the study duration.
Before commencement of dosing, and approximately every 2 months thereafter
for the duration of the study, an ophthalmoscopic examination of all animals
was conducted.
Necropsy and Histopathological Evaluation. All animals were subject to
a complete necropsy including those found dead or moribund. Euthanasia was
performed with a CO2 atmosphere. At the scheduled sacrifices, organ weights
were obtained from a range of tissues including liver, ovaries, uterus, and
spleen. For histopathology, tissue specimens were taken from liver (1 section
from each of 7 lobes), ovaries, oviducts, uterus, mammary gland, adrenals,
pituitary, brain, tail bone, bone marrow (sternum), skin, and all grossly observable lesions, and fixed in 10% neutral buffered formalin. The eyes were
also taken but fixed in Davidson's fixative (26). The carcasses and all other
organs were stored in neutral buffered formalin. Fixed tissues were embedded
in paraffin, sectioned, and stained with Mayer's H&E.
Proliferative liver lesions were diagnosed according to the nomenclature of
Maronpot et al. (27) and semiquantified. Thus, livers at each time interval were
scored for foci of hepatocellular alteration, areas of hepatocellular alteration,
hepatocellular adenomas, and hepatocellular carcinomas. Included in the conventional subgrouping of foci according to tinctorial variation (e.g., clear cell,
basophilic, etc.) was a variant of the basophilic focus where the cytoplasmic
basophilia was arranged in dense linear aggregates, known as a tigroid pattern
(28). In addition, liver sections from the 3-month sacrifice were immunohistochemically stained for the demonstration of the placental form of GST-P as
a marker for preneoplastic foci (29) using the avidin-biotin-peroxidase complex technique (30) and diaminobenzidine for detection of peroxidase binding.
GST-P positive foci were quantitated per cm 2 of liver section by measuring the
surface area of liver tissue on histological slides with a Zeiss Videoplan 2
digital image analysis system.
DNA Adduct Detection by 32p Postlabeling. CrI:CD(BR) rats (3 per
group) were administered toremifene and tamoxifen by gavage in 0.5% carboxymethylcellulose daily for 7 days at dose rates of 48 mg/kg and 45 mg/kg,
respectively. Dose rates 2-fold higher than those used in the chronic toxicity
assay were chosen because of the limited period of exposure in the 32p_
postlabeling study. On day 8, the animals were euthanatized by decapitation
under CO2 sedation and the livers excised for DNA preparation by a modified
chloroform-isoamyl alcohol extraction procedure (31). The isolated DNA was
enzymatically digested into 3'-deoxynucleoside monophosphates. Subsequent
incubation with nuclease P1 cleared the normal nucleosides leaving modified
nucleosides free to be labeled with [32p]ATP. The modified nucleosides were
purified and resolved by thin-layer chromatography and detected with autoradiography (32-34). Hepatic DNA from CrI:CD(BR) rats treated with 60 mg/kg
of DMBA in olive oil (Bertolli USA, Inc., Secaucus, N J) was used as a positive
control for DNA adduct detection. DMBA was purchased from Eastman Kodak
Co. (Rochester, NY) and administered once by gavage to 3 rats, and the livers
harvested 24 h later.
Serum Drug Level Monitoring. During each of the scheduled sacrifices of
the carcinogenicity study, and at the single sacrifice of the postlabeling assay,
blood samples were obtained by cardiac puncture under CO2 anesthesia and
analyzed for achieved serum drug levels. This was carried out by high-performance liquid chromatography after fluorescent activation (35).
Statistical Evaluation. Statistical differences between groups in body and
organ weights and lesion incidences were assessed by either Student's t test or
the Xa test.
Table 1 Group assignment
Scheduled
groups
GLP study
1. Control
2. Toremifene, 12 mg/kg/day
3. Toremifene, 24 mg/kg/day
4. Tamoxifen, 11.3 mg/kg/day
5. Tamoxifen, 22.6 mg/kg/day
Total animals
Pilot study
Toremifene, 48 mg/kg/day
Total
starting
no. of
rats
3
6
12
57
84
75
84
75
9
9
9
9
9
9
9
9
9
9
18
36
10
36
24
15
Total
no. of
unscheduled
deaths
Total
no. of
scheduled
sacrifices
13
20
13
21
9
8
10
34
9
24
49
74
41
75
51
Sacrifice intervals (too.)
and no. of rats sacrificed
290
375
36
10
5
4535
Downloaded from cancerres.aacrjournals.org on June 15, 2017. © 1993 American Association for Cancer
Research.
LIVER EFFECTS OF TOREMIFENE AND TAMOXIFEN
Table 2 Summary of drug and metabolite levels in blood (ng/ml +- SD)
Treatment
(mg/kg)
3 too.
Toremifene
12
24
Tamoxifen
11.3
22.6
6 too.
Toremifene
12
24
Tamoxifen
11.3
22.6
12 mo.
Toremifene
12
24
Tamoxifen
11.3
22.6
Parent
compound
N-Desmethyl
metabolite
4-Hydroxy
metabolite
Total drug
plus metabolite
Metabolite to
drug ratio
59.81 • 81.63
124.93 • 157.10
187.15 • 102.42
446.23 • 254.69
8.18 +-- 12.07
37.58 • 40.72
255.14
608.74
3.27
3.87
146.14 --- 92.71
244.55 • 265.51
88.96 • 39.49
129.94 • 68.19
35.97 • 19.88
47.99 • 25.08
271.07
422.48
0.86
0.73
30.25 • 4.29
53.32 • 58.92
189.46 • 13.75
487.47 • 554.22
17.29 • 0.59
21.79 • 13.16
237.00
562.58
6.84
9.55
71.97 • 9.47
119.66 • 8.66
80.31 • 4.23
132.56 • 14.19
29.87 • 3.55
39.29 • 3.46
182.15
291.51
1.53
1.44
50.32 • 27.35
144.10 • 90.98
206.67 • 87.59
579.92 • 211.48
27.31 • 17.88
56.03 -+ 35.67
284.30
780.05
4.65
4.41
137.84 + 40.65
172.01 • 102.97
136.28 --- 40.78
166.81 • 125.01
56.23 • 6.83
38.44 • 12.75
330.35
377.26
1.40
1.19
RESULTS
Analytical Chemistry and Serum Drug Levels
Concentrations of tamoxifen and toremifene in the dosing suspensions were within +_ 10% of the target levels. These drug preparations
were homogeneous and remained stable for 7 days. Serum drug level
data at 3, 6, and 12 months, shown in Table 2, indicated that for both
tamoxifen and toremifene there was absorption and dose proportionality of the parent compounds and their metabolites at each time
period. The serum levels of parent compound were significantly
higher for tamoxifen than for toremifene, while the levels of metabolites were higher for toremifene. Thus, serum metabolite:drug ratios
were much higher in the toremifene-treated groups, indicating greater
or more rapid metabolic conversion of the parent compound than with
tamoxifen. However, the total serum levels of parent drug plus metabolites were comparable between toremifene and tamoxifen, indicating comparable bioavailability, with a tendency towards a higher
overall exposure to the former drug. With each compound, the major
circulating metabolite was the N-desmethyl metabolite, the serum
concentration of this product being significantly higher for toremifene
than for tamoxifen at each time interval. The serum levels of the
4-hydroxy metabolite were fairly equivalent for each drug.
Mortality
Unscheduled deaths occurred in all treatment groups but were increased in those exposed to the high dose of each drug (Table 1). In
the high-dose toremifene group the deaths were mainly gavage-related
due to increased aggressiveness, which made the animals difficult to
handle. With tamoxifen, mortality was attributed to gavage accidents
due to increased aggressiveness as well as, in the later stages, the
presence of liver tumors.
logical effects known to induce aggressive behavior in rodents (37). A
later observation was the presence of palpable abdominal masses in
tamoxifen-treated groups by 12 months.
A low incidence of drug-related, but not dose-related, alopecia was
more pronounced in tamoxifen- than in toremifene-exposed animals.
High-dose tamoxifen rats in particular exhibited an unthrifty condition
(general wasting and unhealthy coat) related to the presence of liver
tumors.
Cataracts were observed by ophthalmoscopy in 4 rats given tamoxifen, 2 at each dose level. The cataracts were bilateral in 3 of these and
unilateral in 1 low-dose animal. One rat in the high-dose toremifene
group had a unilateral cataract at the 15-month examination i.e., after
the 3-month recovery period.
Postmortem Observations
The most pertinent changes observed at necropsy were the presence
of solitary or multiple liver masses in animals given tamoxifen at the
12- and 15-month sacrifice periods. In many cases, particularly at 15
months, almost all normal liver was replaced with tumor masses (Fig.
2). Compared with controls, other treatment-related changes with both
drugs at necropsy were an increased incidence of cystic ovaries and
decreased size of the uteri.
Among the organ weights taken, a decrease in absolute (but not
relative) liver weights was evident in all treated groups at 3 and 6
months, persisting at 12 months for both toremifene groups. In contrast, the absolute and relative liver weights for both tamoxifen groups
increased significantly at 12 months, becoming more pronounced by
the end of the recovery phase (Table 4). This real increase in liver
weight in rats treated with tamoxifen correlated directly with the
presence of liver masses. Absolute and relative uterine weights were
consistently decreased in all treated groups at each sacrifice point
In-Life Observations
Table 3 Mean body weights of rats at scheduled sacrifices
Results are expressed as means in g • SD.
When compared with controls, all exposed groups in the carcinogenicity study showed significantly lower body weights, commencing
from 5 weeks, continuing throughout the drug treatment period, and
persisting until the end of the recovery phase (Table 3). Reduced body
weight gain is a well recognized effect in female rats treated with
antiestrogens (36). The most significant early clinical observation in
exposed groups was increased aggressiveness, particularly in the highdose toremifene group, probably due to the drug-induced endocrino-
Group
no.
1
2
3
4
5
3 mo.
355.7
242.3
246.8
245.6
249.7
+ 71.7
• 10.2 a
"+ 20.5 a
--- 24.3 a
• 10.4 a
6 mo.
430.3
296.3
302.3
284.1
269.9
+•
•
•
•
116.2
27.1 a
35.8 a
40.4 a
27.4 a
12 mo.
501.9
342.8
340.3
349.0
295.8
+•
•
•
•
95.1
42.3 a
32.2 a
54.6 a
38.1 a
a Statistically different from group 1, P < 0.01.
b Statistically different from group 1, P < 0.05.
4536
Downloaded from cancerres.aacrjournals.org on June 15, 2017. © 1993 American Association for Cancer
Research.
15 mo.
432.3
386.2
376.7
359.0
322.2
--- 105.8
• 48.7
• 64.6
• 43.2 b
• 38.3 a
LIVER EFFECTS OF TOREMIFENE AND TAMOXIFEN
Table 4 Absolute (g) and relative (%) organ weights at 15 months
Results are expressed as means in g or percentage _+ SD.
Group
no.
1
2
3
4
5
Liver
Uterus
Spleen
17.67 _+ 4.10g
4.21 _+ 1.01%
14.36 _+ 2.28 ga
3.72 -+ 0.41%
14.86 - 4.28 g
3.70 _+ 0.39%
30.03 -+ 30.46 g
8.32 _+ 9.34%
54.60 _+ 30.18 g"
18.00 _+ 11.23%`"
0.92 _+ 0.11 g
0.22 _+ 0.06%
0.48 -+ 0.08 a
0.13 -+ 0.03"
0.55 -+ 0.20 g`"
0.15 -+ 0.06%~
0.55 -+ 0.32 ga
0.16 +_ 0.10%"
0.55 _+ 0.20 g`"
0.17 -+ 0.06% b
0.86 - 0.21 g
0.19 +- 0.09%
0.63 -+ 0.13 g`"
0.17 - 0.04%b
0.68 -+ 0.17g b
0.19 -+ 0.07%
0.77 +- 0.30 g
0.22 -+ 0.10%
1.02 -+ 0.24 g
0.33 + 0.10%"
'~ Statistically different from group 1, P < 0.01.
b Statistically different from group 1, P < 0.05.
one animal had a solitary hepatocellular carcinoma (11%). The most
Fig. 2. Liver from the high-dose tamoxifen group showing extensive involvement with
multiple hepatocellular carcinomas.
except 12 months. The only other organ weight change w a s in the
relative spleen weights, w h i c h w e r e increased in the high-dose
tamoxifen group at each sacrifice point.
Histopathological Evaluation
The pertinent pathological changes are described for each involved
organ.
Liver. The incidences o f hepatoproliferative lesions o b s e r v e d at
each o f the 4 sacrifice points are s h o w n in Table 5. At 3 months, the
only lesions present in H & E sections w e r e a low incidence o f solitary
foci o f hepatocellular alteration in the controls (11%) and high-dose
tamoxifen (33%). B y 6 months, the same b a c k g r o u n d o f solitary foci
w a s seen in controls and toremifene groups, but the incidence of
solitary foci in the high-dose tamoxifen group had risen to 89%, and
c o m m o n type of focus in the tamoxifen groups at these early stages
w a s the clear cell or mixed clear/eosinophilic cell type, but basophilic
foci w e r e also present in the high-dose group at 6 months. It is
important to note that no o b v i o u s hepatocyte necrosis, either focal or
single cell death, w a s o b s e r v e d in nonneoplastic liver tissue o f tamoxifen-treated rats at these early time points.
At 12 months, the b a c k g r o u n d incidence o f solitary foci in controls
w a s 94% but statistically lower in the 2 toremifene groups at 5 3 % for
the l o w dose and 2 0 % for the high dose (see Table 5). The l o w e r count
o f foci in the latter groups w a s due to a marked reduction o f tigroid
loci specifically. In the toremifene groups, there were no rats with
multiple foci or areas o f hepatocellular alteration, or a d e n o m a s or
carcinomas, even at the 4 8 - m g / k g dose. In contrast, both tamoxifen
groups had very high incidences o f multiple foci, multiple areas,
adenomas, and carcinomas, each o f these being 100% at the high dose.
Thus, the incidence o f total liver n e o p l a s m s ( a d e n o m a s and carcinomas c o m b i n e d ) w a s 6 7 % for l o w - d o s e tamoxifen and 100% for the
high dose. At the histological level, the t a m o x i f e n - i n d u c e d carcinomas
Table 5 Incidence of hepatocellular proliferative lesions
Foci
Solitary
Areas
Multiple
Solitary
Multiple
Adenomas
Carcinomas
Adenomas and
carcinomas
combined
3 mo.
Group 1
Group 2
Group 3
Group 4
Group 5
1/9 (11)`"
0/9
0/9
0/9
0/9
0/9
0/9
3/9 (33)
0/9
0/9
0/9
0/9
0/9
0/9
0/9
6 too.
Group 1
Group 2
Group 3
Group 4
Group 5
Pilot
1/9 (11)
2/9 (22)
1/9 (11)
3/9 (33)
8/9 b (89)
0/9
0/9
0/9
0/9
0/9
0/9
2/9 (22)
1/9(11)
12 too.
Group 1
Group 2
Group 3
Group 4
Group 5
Pilot
17/18 (94)
19/36b (53)
2/10 ~ (20)
4/36 (11)
0/24
0/18
0/36
0/10
32/36c (89)
24/24 ~ (100)
3/18 (17)
3/36 (8)
1/10 (10)
6/36 (17)
0/24
0/18
0/36
0/10
22/36 c (61)
24/24 ~ (100)
10/13 (77)
13/20 (65)
8/13 (62)
5/21 (24)
0/9
2/13 (15)
0/20
4/13 (31)
1/20a (5)
1/13 (8)
0/20
0/13
14/21 b (67)
9/9 ~ (100)
15 mo.
Group 1
Group 2
Group 3
Group 4
Group 5
0/13
16/21b (76)
9/9 c (100)
0/9
0/9
0/9
0/9
0/9
0/9
0/9
0/9
0/9
0/9
0/9
0/9
0/9
0/9
0/9
0/9
0/9
0/9
0/9
0/9
1/9 (11)
0/9
0/9
0/9
0/9
0/9
0/9
0/9
0/9
0/9
O/lO
0/9
0/9
0/9
0/9
0/9
0/9
0/9
1/9 (11)
O/lO
0/9
0/9
0/13 a
3/21 (14)
0/9
0/18
0/36
0/10
21/36 c (58)
24/24 ~ (100)
0/5
0/13
0/20
0/13
13/21c (62)
9/9 c (100)
0/9
1/9 (11)
O/lO
0/18
0/36
0/10
16/36c (44)
24/24 c (100)
0/5
0/18
0/36
0/10
24/36 ~ (67)
24/24 c (100)
0/5
0/13
0/20
0/13
13/21c (62)
8/9 ~ (89)
0/13
0/20
0/13
15/21 c (71)
9/9 c (100)
`"Numbers in parentheses, percentages.
b Statistically different from group 1 controls, P < 0.01.
c Statistically different from group 1 controls, P < 0.001.
d Statistically different from group 1 controls, P < 0.05.
4537
Downloaded from cancerres.aacrjournals.org on June 15, 2017. © 1993 American Association for Cancer
Research.
LIVER EFFECTS OF TOREMIFENE AND TAMOXIFEN
were typified by trabecular areas of neoplastic hepatocytes, and the
extensive involvement of liver tissue in all lobes was emphasized in
the majority of cases. In one high-dose tamoxifen rat, there was
metastasis of hepatocarcinoma to the lung.
The pattern of lesion development differed little at 15 months after
a 3-month recovery period. Toremifene groups had statistically fewer
solitary areas of hepatocellular alteration than controls, and again, no
liver neoplasms. The incidence of hepatocellular neoplasms in the
low- and high-dose tamoxifen groups was 71% and 100%, respectively, indicating no regression during the treatment-free phase of the
study.
GST-P positive foci were enumerated in livers from the 3-month
sacrifice. The results, summarized in Table 6, demonstrated a statistically significant increase in preneoplastic lesions in the high-dose
tamoxifen group and a statistically significant reduction of foci in the
high-dose toremifene group.
Reproductive Tract. Compound-related effects were observed in
ovaries, oviducts, and uteri, particularly at the 12-month sacrifice,
with the 15-month groups exhibiting a degree of recovery in these
various changes. The ovarian changes differed qualitatively between
toremifene and tamoxifen treatments. Toremifene was associated primarily with an increase in the number of dilated secondary follicles
and some ovarian atrophy. With tamoxifen, the changes were ovarian
atrophy and increased numbers of atretic follicles. Other changes
observed with both drug treatments were increased formation of cysts
derived from either secondary follicles or bursa, and low incidences of
stromal hypercellularity and granulosa cell hyperplasia/tumors (Table
7). Lesions diagnosed as granulosa cell tumor were observed in 1 rat
of each of the treated groups at the 12-month sacrifice. There were
none, however, at 15 months.
Oviduct and uterine changes were evident from 3 months in all
treated groups. The oviducts were typified by vacuole-like holes in the
mucosal lining, possibly representing single cell loss. Uterine changes
involved glandular atrophy.
Mammary Glands. Tamoxifen and toremifene both produced an
acinar atrophy in mammary tissue evident throughout the study. Control rats displayed a low incidence of focal glandular hyperplasia and
mammary adenoma or carcinoma, which was markedly different from
all drug-treated groups in which there were no proliferative lesions of
mammary tissue (Table 7).
Pituitary. At 12 and 15 months, there was a marked difference in
the incidence of pars distalis hyperplasia and pitutary adenoma or
carcinoma between control animals and those treated with either of the
2 drugs (Table 7). No neoplasms were observed in the pituitaries of
drug-treated rats compared with up to a 33% incidence in controls.
Pars distalis hyperplasia was observed in a few toremifene-treated
rats at 15 months, but this was substantially lower than the 50%
incidence in control animals.
Other Tissues. Both tamoxifen and toremifene were associated
with an increase in osteoporotic thinning of trabeculae at the epiphyseal growth plates of tailbone vertebrae at all sacrifice points, although
a recovery trend was detectable at 15 months. Such bone loss is a
Table 6 Incidence of GST-P positive liver loci at 3 months
Group no.
No. of rats with
loci (%)
No. of loci in liver
lobe sections
No. of foci/cm2 of
liver tissue
1
2
3
4
5/9 (56)
3/9 (33)
1/9 a (11)
7/9 (78)
8
7
2
14
0.30
0.34
0.09
0.76
a Statistically different from group 1 controls, P < 0.05.
b Statistically different from group 1 controls, P < 0.01.
reflection of the known antiestrogenic properties of these 2 compounds in the rat (38). Avery low compound-related incidence of fiber
degeneration in the optic lens was seen in tamoxifen-treated rats at 12
and 15 months. No differences were observed in the adrenal glands
between control and treated animals. In the spleens of high-dose
tamoxifen rats, reactive follicular hyperplasia secondary to extensive
liver neoplasia was a common observation, accounting for the statistically significant spleen weight increase in this group.
Unscheduled Deaths
No liver neoplasms were observed in the toremifene-treated animals representing unscheduled deaths, but almost all had lung changes
indicative of gavage accident. In the tamoxifen-treated groups, 1 of 9
(11%) unscheduled deaths had liver neoplasia at the low dose, and 13
of 24 (54%) at the high dose. In the latter group, all but one of the rats
dying at 7.5 months or later had liver tumors.
DNA Base Modification
Typical autoradiograms resulting from the 32p-postlabeling assay of
rat liver after 7 days of drug treatment are presented in Fig. 3. Liver
samples from each animal were examined individually but the results
within groups were uniform. No spots, representing DNA adducts,
were found in any of the vehicle-treated control or toremifene-exposed rat livers. In contrast, 7 spots were identifiable in the autoradiograms of liver cell DNA from the 3 tamoxifen-treated rats, while 5
spots were present in liver DNA from the 3 positive control livers
(DMBA-treated).
DISCUSSION
The most significant findings of the chronic toxicity study were the
confirmation of the potent hepatocarcinogenicity of tamoxifen, and
the lack of any carcinogenic activity of toremifene under the conditions of 12-month daily exposure at high dose in female rats. Tamoxifen-induced liver tumors were multiple and aggressive, leaving little
normal tissue remaining by 15 months. Based additionally on clinical
and necropsy observations, there was progression of the tamoxifeninduced liver tumors during the treatment-free period. In addition, the
main cause of unscheduled death in tamoxifen-treated groups was
liver neoplasia. The present report provides evidence, in compliance
with GLP standards and in test groups of adequate size, for a major
difference in the toxicology of these 2 drugs. The tamoxifen results
concur with those from 2 other studies, which demonstrated liver
tumors using a similar schedule of administration in Sprague-Dawley
rats, but including a higher dose level (19, 20). Consistent with its
hepatocarcinogenicity, tamoxifen induced an increased incidence of
foci of hepatocellular alteration as early as 3 months in our sequential
study. Foci have been established as the precursors of liver neoplasms,
and their induction is predictive of carcinogenic potential (39), as in
this case.
In striking contrast to tamoxifen, toremifene was not only lacking
in hepatocarcinogenic activity at daily doses ranging from 11 to 48
mg/kg but, in addition, this drug reduced the incidence of altered liver
cell loci at the various interim sacrifice stages. This inhibitory effect
on the development of liver foci was evident at 3 months using GST-P
immunoreactivity and at 12 months by conventional H&E staining.
5
The main type of focus suppressed by toremifene was the type designated as having a tigroid pattern of staining (28).
9/9 a (100)
The only proliferative change associated with drug treatment other
86
than hepatocellular involved the ovary, where a very low incidence of
granulosa cell hyperplasia and tumor was observed at 12 months in
3.83 b
each of the tamoxifen and toremifene groups. In the original safety
evaluation of tamoxifen by the manufacturer, granulosa cell tumors
4538
Downloaded from cancerres.aacrjournals.org on June 15, 2017. © 1993 American Association for Cancer
Research.
LIVER EFFECTS OF TOREMIFENEAND TAMOXIFEN
Table 7 Incidence of proliferative lesions in other tissues at 12 and 15 months
12 mo., group:
1
15 mo., group:
2
3
4
5
0/17
0/17
1/34 (3)a
1/34 (3)
0/10
1/10 (10)
3/33 (9)
1/33 (3)
0/24
1/24 (4)
0/13
0/13
4/18 (22)
1/18 (6)
0/36
0/36
0/10
0/10
0/36
0/36
0/24
0/24
Pituitary
Pars distalis hyperplasia
6/18 (33)
Adenoma
4/18 (22)
Carcinoma
0/18
a Numbers in parentheses, percentage.
0/36
0/36
0/36
0/10
0/10
0/10
0/36
0/36
0/36
0/24
0/24
0/24
Ovary
Granulosa cell hyperplasia
Granulosa cell tumor
Mammary
Focal glandular hyperplasia
Adenoma or carcinoma
were recorded in mice (12). The absence of lesions diagnosed as
granulosa cell tumors at 15 months after a 3-month recovery phase in
our study suggests the possibility that this type of ovarian cell proliferation may have represented a reversible hyperplasia.
The 32p-postlabeling study revealed that tamoxifen, but not toremifene, caused accumulation of modified nucleosides in rat liver,
confirming evidence in similar studies from other laboratories that
tamoxifen produces covalent D N A adduct formation in the target
organ of carcinogenicity. Han and Liehr (21), who administered
tamoxifen by i.p. injection to Sprague-Dawley rats at a rate of 20
mg/kg/day, found 2 D N A adducts after a single injection, and 4
additional adducts of increasing intensities at 3 and 6 consecutive days
of treatment. Our study is in agreement in finding 7 adduct spots after
7 days of p.o. administration of a higher dose of tamoxifen. White et
al. (22) also detected 7 adduct spots in Fischer 344 rat D N A following
1
2
3
4
5
0/20
0/20
1/13 (8)
0/13
0/21
0/21
0/9
0/9
2/13 (15)
2/13 (15)
0/20
0/20
0/13
0/13
0/21
0/21
0/9
0/9
6/12 (50)
4/12 (33)
3/12 (25)
1/19 (5)
0/19
0/19
2/12 (17)
0/12
0/12
0/21
0/21
0/21
0/9
0/9
0/9
p.o. gavage with 67.6 mg/kg/day for 7 days. The pattern of spots
found in the latter and in our study appear to be very similar, inlcuding
the relative location of the largest adduct. Tamoxifen has also been
shown to cause D N A adduct formation in C57BL/6 and DBA/2 mouse
strains, but at approximately one-third of the level detected in rat liver
D N A (22). Furthermore, tamoxifen induced unscheduled D N A synthesis in hepatocytes from rats pretreated with the drug, although not
in hepatocytes derived from untreated rats (22). In explanation, White
et al. (22) suggest that the drug may induce its own activating enzyme
system in vivo. T h e same authors have also demonstrated that tamoxifen is positive in the micronucleus test using a metabolically competent human cell line (22), indicating a potential for producing chromosomal anomalies in humans. On the evidence now assembled from
the positive data published on carcinogenicity and short-term assays,
tamoxifen should be regarded as a DNA-reactive agent. On the other
Fig. 3. Autoradiogramsfrom 32p-postlabelingassay of rat liver. DNA from rats treated with tamoxifen (a), toremifene (b), dimethylbenzanthracene
(positive control) (c), and carboxymethylcellulose
vehicle (negative control) (d).
4539
Downloaded from cancerres.aacrjournals.org on June 15, 2017. © 1993 American Association for Cancer
Research.
LIVER EFFECIS OF TOREMIFENE AND TAMOXIFEN
hand, toremifene appears to have no genotoxic properties to date. between rats and humans (44), then the dose level given prophylacUnlike tamoxifen, it lacks hepatocarcinogenic activity in rats, and has tically in women is higher than a hepatocarcinogenic dose in rats.
been shown in our study with the Sprague-Dawley strain, and by Consequently, the margin of safety would seem insufficient to justify
White et al. (22) with the Fischer 344, not to produce DNA adducts in the long-term use of this drug in relatively young cancer-free women
rat liver.
unless it can be proven that the rat data have no human relevance.
In conclusion, our study has shown that tamoxifen is strongly
Considering the minor difference in molecular structure between
the 2 drugs, the strong hepatocarcinogenicity of tamoxifen, contrast- hepatocarcinogenic in female rats, with DNA adduct forming ability
ing with a lack of hepatocarcinogenicity with toremifene, is striking. indicative of a DNA-reactive carcinogen. The potency of its carcinoThe basis for this difference in biological effect is not known, but we genic action coupled with DNA damaging propensity indicate that
suggest that the carcinogenicity of tamoxifen may be due to activation tamoxifen has tumor initiating activity for the liver. In contrast, a close
at the ethylene bond region of the molecule, as has been proposed for sructural analogue with very similar antiestrogenic properties, torediethylstilbestrol (40). This is in accordance with the finding that mifene, was without hepatocarcinogenicity or DNA modifying effects.
diethylstilbestrol is also hepatocarcinogenic in female rats and pro- Like tamoxifen, toremifene suppressed spontaneous tumor formation
duces DNA alterations in rat liver (41). Such a hypothesis raises the in endocrine-regulated organs, specifically mammary and pituitary
possibility that toremifene has a lower capacity for generation of glands, a likely result of antihormonal effects. In addition, the inhigenotoxic metabolites because of steric hindrance by the presence of bition of liver foci by toremifene suggests that this drug may have
a bulky chlorine atom adjacent to the ethylene double bond. Certainly, anticarcinogenic activity for liver as well.
the carcinogenic action of tamoxifen cannot be ascribed solely to its
hormonal activity because toremifene exerts qualitatively and quan- ACKNOWLEDGMENTS
titatively similar hormone-related effects on female rat liver as
The authors gratefully acknowledge Dr. Mirjana Djordjevic of the American
tamoxifen (1, 2). In the absence of any conspicuous focal or single cell
Health Foundation for preparation of the test articles, and Dr. Marjorie
necrosis in nonneoplastic liver tissue, tamoxifen-induced hepatocarNeaderland, Diplomate, American College of Veterinary Ophthalmologists,
cinogenicity cannot be linked to cytotoxicity. Conversely, the nonex- Animal Eye Clinic, Norwalk, CT, for performing the ophthalmological examiistent hepatocarcinogenicity of toremifene is not due to any lack of nations.
biological activity in the liver. The difference in hepatocarcinogenicity
is not explained by any differences in serum drug levels, as total REFERENCES
drug/metabolite concentrations of the 2 test compounds were compa1. Kangas, L. Review of the pharmacological properties of toremifene. J. Steroid Biorable and the bioavailability of toremifene appeared to be higher than
chem., 36: 191-195, 1990.
2. Kendall, M. E., and Rose, D. P. The effects of diethylstilbestrol, tamoxifen, and
for tamoxifen. As observed here, toremifene is extensively metabotoremifene on estrogen-inducible hepatic proteins and estrogen receptor proteins in
lized to N-desmethyltoremifene in the rat, but also in humans (13, 42).
female rats. Toxicol. Appl. Pharmacol., 114: 127-131, 1992.
Consequently, higher breast cancer therapeutic levels are required for
3. Jordan, V. C., and Murphy, C. S. Endocrine pharmacology of antiestrogens as antitumor agents. Endrocrine Rev., 11: 578-610, 1990.
toremifene than for tamoxifen because the antitumor properties of
4. Riley, D., Baum, M., MacIntyre, J., Berstock, D., McKinna, A., Jackson, I., Sainsbury,
N-desmethyltoremifene are slightly weaker than for the parent comJ. R. C., Wilson, A., Wheeler, T., Dobbs, J., Rees, G., Powles, T., Rubens, R.,
pound (13). This variation between the 2 drugs provides no basis for
Haybittle, J., McPherson, K., and Houghton, J. The effect of adjuvant tamoxifen: the
latest results from the Cancer Research Campaign adjuvant breast trial. Eur. J. Cancer,
a difference in hepatocarcinogenicity, because toremifene proved
28A: 904-907, 1992.
negative for liver pathology at a dose twice as high as that which
5. Cuzick, J., Wang, D. Y., and Bulbrook, R. D. The prevention of breast cancer. Lancet,
1: 83-86, 1986.
induced 100% incidence of liver cancer by 12 months in tamoxifen6. Nayfield, S. G., Karp, J. E., Ford, L. G., Dorr, E A., and Kramer, B. S. Potential role
treated rats.
of tamoxifen in prevention of breast cancer. J. Natl. Cancer Inst., 83: 1450-1459,
The hepatocarcinogenicity of tamoxifen must be placed in com1991.
7. Jordan, V. C. The strategic use of antiestrogens to control the development and growth
parative pharmacokinetic perspective because of its medical indicaof breast cancer. Cancer (Phila.), 70: 977-982, 1992.
tions in humans. Our concern regarding the cancer prophylactic ap8. Powles, T. J. The case for clinical trials of tamoxifen for prevention of breast cancer.
Lancet, 340: 1145-1147, 1992.
plication has been expressed recently (43). A no-observable-effect
9. Henderson, B. E., Ross, R. K., and Pike, M. C. Hormonal chemoprevention of cancer
level for hepatocarcinogenicity was not established in the present
in women. Science (Washington DC), 259: 633-638, 1993.
study because both exposure levels were associated with liver cancer. 10. Fugh-Berman, A., and Epstein, S. Tamoxifen: disease prevention or disease substitution? Lancet, 340: 1143-1145, 1992.
Notwithstanding, it is relevant to compare the hepatocarcinogenic
11. Powles, T. J., Tillyer, C. R., Jones, A. L., Ashley, S. E., Treleaven, J., Davey, J. B., and
dose of tamoxifen in rats with the doses administered to humans. For
McKinna, J. A. Prevention of breast cancer with tamoxifen--an update on the Royal
adjuvant therapy of early-stage breast cancer, the standard dose is 20
Marsden Hospital pilot programme. Eur. J. Cancer, 26: 680-684, 1990.
rag/day (6), which is the same as the daily total used long-term in 12. E. R. Barnhart (ed.). ICI Pharma Nolvadex. In: Physicians Desk Reference, Ed. 45,
pp. 1070-1072. Oradell, NJ: Medical Economics, Inc., 1991.
national trials of tamoxifen as a chemoprophylactic agent in women at 13. Kangas, L. Development and Biochemical Pharmacology of Toremifene, an Antiestrogenic Anticancer Drug, pp. 1-88. Turku, Finland: Farmos Group, Ltd., 1990.
high risk of developing breast cancer (11). Using a scaling factor for
body surface of 37 for a 60-kg woman, the therapeutic and prophy- 14. Kallio, S., Kangas, L., Blanco, G., Johansson, R., Karjalainen, R., Perila, M., Pippo,
I., Sundquist, H., SodervaU, M., and Toivola, R. A new triphenylethylene compound,
lactic doses of the drug convert to 12.3 mg/m2/day. Extrapolating from
Fc-1157a. I. Hormonal effects. Cancer Chemother. Pharmacol., 17: 103-108, 1986.
15. Valavaara, R., Pyrh6nen, S., Heikkinen, M., Rissanen, P., Blanco, G., Th61ix, E.,
the rat study, the lowest hepatocarcinogenic dose of tamoxifen (11.3
Nordman, E., Taskinen, P., Holsti, L., and Hajba, A. Toremifene, a new antiestrogenic
mg/kg/day), converted to 73.5 mg/m2/day using a scaling factor of 6.5
compound, for treatment of advanced breast cancer. Phase II study. Eur. J. Cancer
for the rat, is 6 times higher than the standard therapeutic and proClin. Oncol., 24: 785-790, 1988.
phylactic doses administered to women. Taking this a step further to 16. Gundersen, S. Toremifene, a new antiestrogenic compound in the treatment of metastatic mammary cancer. A phase II study. J. Steroid Biochem. 36: 233-234, 1990.
account for internal drug exposure, our data show that the mean serum
17. Kaufman, M., Possinger, K., Illiger, H. J., Schmid, H., Hietanen, T., Johansson, R.,
Pyrhonen, S., Valavaara, R., and Sindermann, H. Toremifene: clinical phase II/III
levels of tamoxifen in the ll.3-mg/kg group of rats ranged from about
trials of a new antiestrogen in patients with advanced breast cancer. Contrib. Oncol.,
72 to 146 ng/ml between 3 and 12 months of treatment. This range
37: 50-57, 1989.
appears to be comparable to the 125 ng/ml of tamoxifen found by
18. Pyrhonen, S., Phase III studies of toremifene in metastatic breast cancer. Breast
Cancer Res. Treat., 16: 41-46, 1990.
Robinson et al. (42) in the serum of women receiving 10 mg twice
19. Williams, G. M., Iatropoulos, M. J., Djordjevic, M. V., and Kaltenberg, O. P. The
daily, or 12.3 mg/m2/day. If a factor of 6 is accepted to account for the
triphenylethylene drug tamoxifen is a strong liver carcinogen in the rat. Carcinogencombined differences in liver tissue volume and liver plasma flow rate
esis (Lond.), 14: 315-317, 1993.
4540
Downloaded from cancerres.aacrjournals.org on June 15, 2017. © 1993 American Association for Cancer
Research.
LIVER EFFECTS OF TOREMIFENE AND TAMOXIFEN
20. Hirsimaki, E, Hirsimaki, Y., Nieminen, L., and Payne, B. J. Tamoxifen induces
hepatocellular carcinoma in rat liver: a 1-year study with two antiestrogens. Arch.
Toxicol., 67: 49-54, 1993.
21. Han, Y., and Liehr, J. G. Induction of covalent DNA adducts in rodents by tamoxifen.
Cancer Res., 52" 1360--1363, 1992.
22. White, I. N. H., de Matteis, E, Davies, A., Smith, L. L., Crofton-Sleigh, C., Venitt, S.,
Hewer, A., and Phillips, D. H. Genotoxic potential of tamoxifen and analogues in
female Fischer F344/n rats, DBA/2 and C57BL16 mice and in human MCL-5 cells.
Carcinogenesis (Lond.), 13: 2197-2203, 1992.
23. Reddy, M. V., and Randerath, K. Nuclease Pl-mediated enhancement 32P-postlabeling
test for structurally diverse DNA adducts. Carcinogenesis (Lond.), 7: 1543-1551,
1986.
24. National Toxicology Program. General Statement of Work for the Conduct of Toxicity
and Carcinogenicity Studies in Laboratory Animals, Appendix H, p. 5. April 1987.
25. Rao, G. N., and Knapka, J. J. Contaminant and nutrient concentrations of natural
ingredient rat and mouse diet used in chemical toxicology studies. Fund. Appl.
Toxicol., 9: 329-338, 1987.
26. Toyoda, K., Furukawa, E, Hasegawa, R., Sato, H., Jang, J. J., Takahashi, M., and
Hayashi, Y. The diagnostic value of Davidson fixation method on toxicological
evaluation of eye lesion--histopathological findings of cataract formation following
galactose diet intake in rats (in Japanese). Bull. Natl. Inst. Hygienic Sciences (Tokyo),
104: 82-86, 1986.
27. Maronpot, R. R., Montgomery, C. A., Boorman, G. A., and McConnell, E. E. National
Toxicology Program nomenclature for hepatoproliferative lesions of rats. Toxicol
Pathol., 14: 263-273, 1986.
28. Bannasch, P., Benner, U., Enzman, H., and Hacker, H. J. Tigroid cell foci and
neoplastic nodules in the liver of rats treated with a single dose of aflatoxin B1.
Carcinogenesis (Lond.), 6: 1641-1648, 1985.
29. Sato, K., Kitahara, A., Satoh, K., Ishikawa, T., Tatematsu, M., and Ito, N. The
placental form of glutathione S-transferase as a new marker protein for preneoplasia
in rat chemical hepatocarcinogenesis. Jpn. J. Cancer Res., 75: 199-202, 1984.
30. Hsu, S-M., Raine, L., and Fanger, H. Use of avidin-biotin-peroxidase complex (ABC)
in immunoperoxidase techniques: a comparison between ABC and unlabeled antibody
(PAP) procedures. J. Histochem. Cytochem., 29: 577-580, 1981.
31. Marmur, J. A. Procedure for the isolation of deoxyribonucleic acid from microorganisms. J. Mol. Biol., 3: 208-218, 1961.
32. Gupta, R. C., Reddy, M. V., and Randerath, K. 32p-postlabeling analysis of nonradioactive aromatic carcinogen-DNA adducts. Carcinogenesis (Lond.), 3: 10811092, 1982.
33. Gupta, R. C. Enhanced sensitivity of 32p-postlabeling analysis of aromatic carcinogen:DNA adducts. Cancer Res., 45: 5656--5662, 1985.
34. Gupta, R. C. 32P-postlabeling assay to measure carcinogen-DNA adducts. Prog. Exp.
Tumor Res., 31: 21-32, 1987.
35. Holleran, W. M., Gharbo, S. A., and DeGregorio, M. W., Quantitation of toremifene
and its major metabolites in human plasma by high-performance liquid chromatography following fluorescent activation. Anal. Lett., 20: 871-879, 1987.
36. Perry, B. N., McCracken, A., Furr, B. J., and MacFie, H. J. Separate roles of androgen
and oestrogen in the manipulation of growth and efficiency of food utilization in
female rats. J. Endrocrinol., 81: 35-48, 1979.
37. Sigg, E. B., Day, C., and Colombo, C. Endocrine factors in isolation-induced aggressiveness in rodents. Endocrinology, 78: 679-684, 1966.
38. Feldmann, S., Minne, H. W., Parvizi, S., Pfeifer, M., Lempert, U. G., Bauss, E, and
Ziegler, R. Antiestrogen and antiandrogen administration reduce bone mass in the rat.
Bone Miner., 7." 245-254, 1989.
39. Williams, G. M. The significance of chemically induced hepatocellular altered foci in
rat liver and application to carcinogen detection. Toxicol. Pathol., 17: 663-674, 1989.
40. Metzler, M., and Degen, G. H. Sex hormones and neoplasia: liver tumors in rodents.
Arch. Toxicol. Suppl., 10: 251-263, 1987.
41. Williams, G. M., Iatropoulos, M., Cheung, R., Radi, L., and Wang, C. X. Diethylstilbestrol liver carcinogenicity and modification of DNA in rats. Cancer Lett., 68:
193-198, 1993.
42. Robinson, S. P., Langan-Fahey, L., Johnson, D. A., and Jordan, V. C. Metabolites,
pharmacodynamics and pharmacokinetics of tamoxifen in rats and mice compared to
the breast cancer patient. Drug Metab. Dispos., 19: 36-43, 1991.
43. Williams, G. M., latropoulos, M. J., and Hard, G. C. Long-term prophylactic use of
tamoxifen: is it safe? Eur. J. Cancer Prevention, 1: 386--387, 1992.
44. Iatropoulos, M. J. Accelerated rodent bioassay predictive of chemical carcinogenesis.
Exp. Toxicol. Pathol., 44: 481-487, 1992.
4541
Downloaded from cancerres.aacrjournals.org on June 15, 2017. © 1993 American Association for Cancer
Research.
Major Difference in the Hepatocarcinogenicity and DNA
Adduct Forming Ability between Toremifene and Tamoxifen in
Female Crl:CD(BR) Rats
Gordon C. Hard, Michael J. Iatropoulos, Kevin Jordan, et al.
Cancer Res 1993;53:4534-4541.
Updated version
E-mail alerts
Reprints and
Subscriptions
Permissions
Access the most recent version of this article at:
http://cancerres.aacrjournals.org/content/53/19/4534
Sign up to receive free email-alerts related to this article or journal.
To order reprints of this article or to subscribe to the journal, contact the AACR Publications
Department at [email protected].
To request permission to re-use all or part of this article, contact the AACR Publications
Department at [email protected].
Downloaded from cancerres.aacrjournals.org on June 15, 2017. © 1993 American Association for Cancer
Research.