Chlordecone Exposure and Risk of Prostate Cancer
Luc Multigner, Jean Rodrigue Ndong, Arnaud Giusti, Marc Romana, Helene Delacroix-Maillard,
Sylvaine Cordier, Bernard Je´gou, Jean Pierre Thome, and Pascal Blanchet
From the Institut National de la Sante´
et de la Recherche Me´ dicale (Inserm) U
625, Pointe a` Pitre; Inserm U 763;
Centre d’Examens de Sante´ Sainte
Genevie` ve; Service d’Urologie, Centre
Hospitalier Universitaire de la Guade-
loupe; Universite´ des Antilles et de la
Guyane, Pointe a` Pitre, Guadeloupe,
French West Indies; Inserm U 625,
Rennes; Universite´ Rennes 1, Rennes,
France; and the Laboratoire d’Ecologie
Animale et d’Ecotoxicologie, Center for
Analytical and Research Technology,
Universite´ de Lie` ge, Lie` ge, Belgium.
Submitted November 28, 2009;
accepted May 4, 2010; published online
ahead of print at www.jco.org on June
21, 2010.
Supported by the Programme Hospi-
talier de Recherche Clinique, the Plan
Pluri-Formation from the French Minis-
try of Education and Research, the
French Ministry of Overseas Territories,
the French Agency for Environmental
and Occupational Health Safety, the
Guadeloupean Division of Social,
Health, and Welfare Affairs Directorate,
the National Health Directorate, the
Association pour la Recherche sur le
Cancer, the Comite´ Guadeloupe Ligue
contre le Cancer; Grant No. 212844
from the European Union (Developmen-
tal Effects of Environment on Repro-
ductive Health).
Authors’ disclosures of potential con-
flicts of interest and author contribu-
tions are found at the end of this
article.
Corresponding author: Luc Multigner,
MD, Institut National de la Sante´etde
la Recherche Me´ dicale U625, Faculte´
de Me´ decine, Campus de Fouillole,
BP145, 97154 Pointe a` Pitre, Guade-
loupe, French West Indies; e-mail:
© 2010 by American Society of Clinical
Oncology
0732-183X/10/2821-3457/$20.00
DOI: 10.1200/JCO.2009.27.2153
ABSTRACT
Purpose
Determining whether environmental estrogens are associated with the risk of prostate cancer may
have important implications for our general understanding of this disease. The estrogenic
insecticide chlordecone was used extensively in the French West Indies, contaminating the
population for more than 30 years. We analyzed the relationship between exposure to chlordecone
and the risk of prostate cancer.
Patients and Methods
We investigated 623 men with prostate cancer and 671 controls. Exposure was analyzed
according to case-control status, using either current plasma concentration or a cumulative
exposure index based on years of exposure. We genotyped two single-nucleotide polymorphisms
(rs3829125 and rs17134592) in the gene encoding chlordecone reductase.
Results
We found a significant increase in the risk of prostate cancer with increasing plasma chlordecone
concentration (odds ratio [OR], 1.77; 95% CI, 1.21 to 2.58 for the highest tertile of values above
the limit of detection [LD]; P trend .002) and for cumulative exposure index (OR, 1.73; 95% CI,
1.04 to 2.88 for the highest quartile; P trend .004). Stronger associations were observed among
those with a positive family history of prostate cancer and among those who had lived in a
Western country. The rs3829125 and rs17134592 allele variants were in complete linkage
disequilibrium and were found at low frequency (0.04). Among subjects with plasma chlordecone
concentrations above the LD, carriers of the allele variants had a higher risk of prostate cancer (OR,
5.23; 95% CI, 0.82 to 33.32).
Conclusion
These findings support the hypothesis that exposure to environmental estrogens increases the
risk of prostate cancer.
J Clin Oncol 28:3457-3462. © 2010 by American Society of Clinical Oncology
INTRODUCTION
Prostate cancer is the most commonly diagnosed
cancer among men in developed countries. How-
ever, little is known about the risk factors associ-
ated with this cancer. Advancing age, ethnic
origins, and a family history of prostate cancer are
the only established risk factors.
1,2
Many lifestyle-
related risk factors have been implicated, but their
roles in prostate cancer etiology remain unclear.
1,2
The effect of exposure to synthetic chemicals in the
environment on prostate cancer development re-
mains a matter of debate with implications for pub-
lic health. Chemicals with hormonal properties, also
called endocrine disruptors, are thought to be in-
volved.
3,4
However, no epidemiologic evidence of a
positive link between environmental exposure to
endocrine chemicals and prostate cancer has yet
been established.
Chlordecone (also known as Kepone) is an or-
ganochlorine insecticide with well defined estro-
genic properties.
5,6
It was extensively used from
1973 to 1993 in the French West Indies, to control
the banana root borer. A few years after the intro-
duction of chlordecone, the widespread pollution of
soils, river water, wild animals, and vegetables grow-
ing in polluted soils was reported.
7,8
This pesticide
undergoes no significant biotic or abiotic degrada-
tion in the environment, so permanently polluted
soils and waters have remained the primary source
of foodstuffs contamination, and human beings
continue to be exposed to this chemical.
9-13
Chlordecone is a potential carcinogen and has
been shown to cause hepatic tumors in laboratory
rats and mice.
14
The carcinogenic and hormonal
properties of chlordecone and its long biologic half-
life raise the possibility of long-term effects, such as
cancer. We tested the hypothesis that chlordecone
JOURNAL OF CLINICAL ONCOLOGY
ORIGINAL REPORT
VOLUME 28 NUMBER 21 JULY 20 2010
© 2010 by American Society of Clinical Oncology
3457
Downloaded from ascopubs.org by 37.58.186.129 on February 8, 2017 from 037.058.186.129
Copyright © 2017 American Society of Clinical Oncology. All rights reserved.
exposure in adulthood, over a 30-year period, favors the development
of prostate cancer in the French West Indies, where the incidence of
this disease is particularly high.
15
Chlordecone reductase catalyzes
the reduction of chlordecone to chlordecone alcohol, increasing the
biliary excretion of chlordecone and decreasing its toxicity.
16
We
therefore hypothesized that the prostate cancer-causing effects of
chlordecone would be stronger in individuals with functional variants
of the chlordecone reductase gene, rs3829125 and rs17134592, result-
ing in the production of a protein with low levels of enzymatic activity.
We report the results of a case-control study in which chlorde-
cone exposure was evaluated by measuring its concentration in
the blood.
PATIENTS AND METHODS
Study Population
This study was conducted in Guadeloupe, a French archipelago in the
Caribbean, most of the 405,000 inhabitants of which are of African descent.
This study was carried out on 709 consecutive incident cases of histologically
confirmed prostate cancer (June 2004 to December 2007) and 723 controls
without prostate cancer (January 2005 to December 2006). For details of the
selection of cases and controls, see the Appendix (online only).
Trained nurses obtained information for both patients and controls,
concerning demographic characteristics, anthropometrics, periods and places
of residence since birth, lifestyle, occupational data, family history of prostate
cancer, medical background, use of medication, and prostate cancer screening
history. Weight, height, and the circumferences of the waist and hips were
measured at the end of the face-to-face interview. Participants were also asked
to provide a blood sample. The study was approved by the Guadeloupean
ethics committee for studies involving human subjects. Each participant pro-
vided written informed consent.
Laboratory Procedures and Data Management and
Statistical Analyses
Chlordecone analysis, total lipid determination, genotyping methods
and linkage analysis are described in the Appendix (online only).
A reference date was assigned to each participant, corresponding to the
date of histopathologic diagnosis of prostate cancer for patients and the last
date of prostate-specific antigen (PSA) determination for controls. Plasma
chlordecone concentrations were classified according to their distribution
among control samples. Values equal to or below the limit of detection (LD)
were used as the reference group and values above the LD were grouped into
tertiles. Based on the realistic assumption that the Guadeloupean population
has been continuously exposed from 1973 to the present day, we obtained a
cumulative exposure index score for each subject. This score was calculated as
the product of current plasma chlordecone concentration and the number of
years of residence in the French West Indies between 1973 and the year of the
reference date. We calculated such scores for subjects with plasma chlordecone
concentrations above LD. The cumulative exposure scores were grouped into
quartiles, based on their distribution among controls, the lowest quartile being
the reference group.
Baseline covariates were compared between patients and controls (Table
1). Odds ratios (OR) and 95% CIs for the risk of prostate cancer were estimated
by unconditional logistic regression. We investigated whether covariates were
confounding factors or effect modifiers, by looking at the association between
chlordecone exposure and prostate cancer separately for each of the following
covariates: age, Caribbean origin, education, body mass index, waist to hip
ratio, alcohol use, smoking, past urogenital infections, diabetes (type 2), viral
infections, prostate cancer screening history (during the last 5 years), family
history of prostate cancer (first degree), banana farming, past residence in
Western countries, year of reference date, total plasma lipid concentration,
and the series and batch number of chlordecone analysis. Missing data for
covariates varied from none to lower than 5%, except for waist to hip ratio and
genotype, for which 17% and 8.6%, respectively, of the data were missing.
Categorical covariates, including missing data as an additional response, were
modeled as dummy variables representing the different levels. Confounding
covariates were included into the logistic model if they modified the OR
estimates by more than 10%. Tests for linear trend across exposure categories
were performed, with the natural log-transformed chlordecone concentration
treated as a continuous variable. For values equal to or below the LD, we
assigned a value of the LD divided by 2. We tested for a modifier effect, by
creating an interaction term based on the cross product of these variables and
exposure categories (treated as a continuous variable). Interactions were ex-
amined by entering the interaction terms into the logistic regression model.
We quantified the significance of the interaction term using the Wald test. We
used polytomous logistic regression to estimate risk simultaneously among
controls and non ordered subgroups of cases, as a function of age at onset
( 60 v 60) and prostate cancer stage (local v regional/distant stage). We also
used a composite index of prostate cancer aggressiveness at diagnosis: high
aggressive potential (PSA 30 ng/mL or regional/distant stage or Gleason
score of 4 3 and higher) and low aggressive potential (PSA 30 ng/mL, local
stage, and Gleason score of 3 4 and lower).
17
P values lower than .05 were
considered significant. All statistical tests were two tailed and were carried out
with SAS version 9.1 (SAS Institute Inc, Cary, NC).
RESULTS
The results presented here were obtained from a study population
comprising 623 of the 709 eligible prostate cancer cases and 671 of the
eligible 722 controls, from whom we were able to obtain blood sam-
ples and determinations of plasma chlordecone concentration. The
baseline characteristics of the study participants are summarized in
Table 1. Chlordecone was detected in the plasma of 68.7% of the cases
and 66.8% of the controls.
Age-adjusted (5-year intervals) and multivariable-adjusted ORs
and 95% CI for the association between prostate cancer and plasma
chlordecone concentrations are presented in Table 2. Using plasma
chlordecone concentration as a means of measuring exposure, we
found that the highest exposure categories were associated with a
significantly higher risk. Moreover, the relationship between exposure
and response was significant (P .002). When we used the cumula-
tive exposure index as a means of measuring exposure, we found that
the highest quartile was associated with a significantly higher risk of
prostate cancer and that the exposure-response relationship was sig-
nificant (P .004).
When we looked for potential interactions, no modifier effects
were observed for any covariates, other than a history of prostate
cancer in first-degree relatives and previous residence in a Western
country. Results for analyses stratified for these two covariates are
presented in Table 3. Associations between chlordecone exposure and
prostate cancer were stronger in men with a family history of prostate
cancer and in men who had previously lived in Western countries,
with significant linear relationships (P .03 and P .005, respec-
tively). The interaction terms were found to be significant for family
history of prostate cancer and past residence in a Western country
(Table 3). Using chlordecone concentrations below the LD as the
reference category and concentrations above LD as the exposure cat-
egory, we carried out a double stratification, according to family his-
tory of prostate cancer and past residence in Western countries.
Subjects with both a family history of prostate cancer and previous
residence in a Western country had a higher risk of prostate cancer
(OR, 4.94; 95% CI, 1.15 to 21.23). The risk did not differ from one for
those with neither or only one of these factors (data not shown).
Multigner et al
3458 © 2010 by American Society of Clinical Oncology
J
OURNAL OF CLINICAL ONCOLOGY
Downloaded from ascopubs.org by 37.58.186.129 on February 8, 2017 from 037.058.186.129
Copyright © 2017 American Society of Clinical Oncology. All rights reserved.
We determined whether the chlordecone-prostate cancer associ-
ation depended on clinical characteristics. We found a significantly
higher risk for patients age 60 years or older (for the highest tertiles of
detectable values, OR, 1.91; 95% CI, 1.24 to 2.94; P for trend .001)
whereas, for patients younger than 60 years, the OR was 1.22 (95%
CI, 0.58 to 2.57; P for trend .94). We found that the risk was
significantly higher for both local (OR, 1.58; 95% CI, 1.07 to 2.35;
P for trend .021) and regional/distant stage (OR, 2.25; 95% CI,
Table 1. Baseline Characteristics of Patients and Controls
Characteristic
Patients (n 623) Controls (n 671)
P
No./Total No. % No./Total No. %
Median age, years 66.2 60.6 .01
Interquartile range 60.5-71.5 54.0-67.1
Caribbean origin .01
French West Indies (Guadeloupe or Martinique) 96.5 91.4
Haiti or Dominica 3.5 8.6
Education .03
High school and higher 13.3 10.7
Secondary 25.4 31.9
Primary 61.4 57.4
Body mass index† .49
Normal ( 25) 44.0 47.0
Overweight (25-29.9) 44.3 41.0
Obese ( 30) 11.7 12.0
Waist-to-hip ratio 0.95 45.4 30.1 .01
Current or past alcohol use 86.8 82.9 .05
Current or past smoking 38.3 37.1 .69
Past urogenital infection 16.1 17.2 .65
Type 2 diabetes 18.1 12.3 .01
Viral infection 23.5 24.1 .66
Prostate cancer screening history‡ 50.7 13.4 .01
Family history of prostate cancer
No 55.9 78.0 .01
Yes 24.4 10.2
Do not know 19.7 11.8
Banana farming 11.9 10.0 .27
Past residence in Western countries, 1 year§ 29.9 23.9 .02
Median duration of residence in Western countries§ 15.1 13.2 .04
Interquartile range 7.0-26.3 6.5-24.8
Median years of residence in French West Indies from 1973 to reference date 32.8 32.7 .43
Interquartile range 31.5-33.8 30.0-33.3
Median plasma chlordecone,
g/L 0.44 0.40 .39
Interquartile range 0.25-1.00 0.25-0.86
Chlordecone reductase SNP (No./total No. with data)
rs3829192
Wild-type C 1,137/1,182 96.2 1,161/1,208 96.1 .91
Variant allele G 45/1,182 3.8 47/1,208 3.9
rs17134592
Wild-type C 1,137/1,182 96.2 1,161/1,208 96.1 .91
Variant allele G 45/1,182 3.8 47/1,208 3.9
Median PSA levels, ng/mL 8.75
Interquartile range 6.00-14.30
Gleason score
7(3 4) 82.0
4 3 and 7 18.0
TNM
T1c or T2 and N0 and M0 13.8
T3 or T4, or N or M 86.2
Abbreviations: SNP, single nucleotide polymorphism; PSA, prostate-specific antigen.
P values for continuous variables are those for non parametric Mann-Whitney rank tests; for plasma chlordecone concentration values equal to or below the limit
of detection, we assigned rank values corresponding to the value of the limit of detection divided by 2; for categorical variables, P values were obtained in tests for
heterogeneity across levels and for SNPs, in tests for Hardy–Weinberg equilibrium among controls.
†The body mass index is the weight of the subject in kilograms divided by the square of the height in meters.
‡Within the 5-year period before the PSA test preceding the histological diagnosis of cases or allowing the selection of controls.
§All but four of the migrants had lived in mainland France. The remaining four migrants had lived in Germany.
Chlordecone Exposure and Prostate Cancer
www.jco.org © 2010 by American Society of Clinical Oncology 3459
Downloaded from ascopubs.org by 37.58.186.129 on February 8, 2017 from 037.058.186.129
Copyright © 2017 American Society of Clinical Oncology. All rights reserved.
1.16 to 4.34; P for trend .018). However, using the composite
index of aggressiveness, a significantly higher risk was found only
for high aggressive forms (OR, 2.16; 95% CI, 1.33 to 3.51; P for
trend .004), with the OR for low aggressive forms being only 1.45
(95% CI, 0.96 to 2.19; P for trend .04).
Both SNPs, rs3829125 and rs17134592, were in Hardy-Weinberg
equilibrium in the controls (P .05). These variants were in complete
linkage disequilibrium with D’ and r
2
of 1, and were present at identi-
cal frequencies in cases and controls. The frequencies of the allele
variants were low (0.04). We therefore stratified the analysis according
to genotype, using chlordecone concentrations at or below the LD as
the reference category and concentrations above the LD as the expo-
sure category. For carriers of variant alleles, a non significant increase
in prostate cancer risk was observed (OR, 5.23; 95% CI, 0.82 to 33.32),
whereas the OR for wild-type allele homozygotes was 1.30 (95% CI,
0.91 to 1.85).
DISCUSSION
This study reveals that chlordecone exposure, evaluated by determin-
ing plasma chlordecone concentration, is consistently associated with
an increase in the risk of prostate cancer, and with a significant
exposure-response relationship. This is the largest study to have inves-
tigated the effects of organochlorine compounds on prostate cancer
risk through the evaluation of exposure by biologic measurement.
Previous studies have been exploratory and based on less than 80
incident or prevalent prostate cancer cases.
18-21
A significant increase in risk was observed after adjustment for
covariates. This unusual finding is explained by the contribution of
waist-to-hip ratio (abdominal obesity) and prostate cancer screening,
which act as negative confounding factors. In our population study,
both abdominal obesity and prostate cancer screening were associated
with a significant increase in the risk of prostate cancer (data not
shown), as reported for abdominal obesity in other populations,
22,23
and for prostate cancer screening. We also found that these two co-
variates were associated with lower plasma chlordecone concentra-
tions in our study. In the case of abdominal obesity, peripheral fat may
sequester organochlorine lipophilic compounds, leading to an inverse
association between peripheral fat levels and circulating concentra-
tions of organochlorine compounds.
24
Many factors may account for
the observed inverse relationship between prostate cancer screening
and plasma chlordecone concentration. For example, we found that
individuals with a history of prostate cancer screening were more
likely to have lived in Western countries, and were less likely to have
worked on banana plantations, these factors in turn being associated
with lower plasma chlordecone concentration. In addition, screening
procedures may introduce distortions in the associations between
exposures of interest and cancer outcomes if the study would have
included fewer cases in the absence of screening.
25
This is particularly
true for Guadeloupe, where the recorded incidence of prostate cancer
Table 2. Plasma Chlordecone Concentration and Risk of Prostate Cancer
Chlordecone No. of Patients No. of Controls
Age Adjusted Multivariable
OR 95% CI OR 95% CI
Plasma concentration,
g/L
0.25 (LD) 195 223 1.00 1.00
0.25-0.47 128 150 0.95 0.69 to 1.31 1.11 0.75 to 1.65
0.47-0.96 139 149 1.16 0.84 to 1.59 1.22 0.82 to 1.83
0.96 161 149 1.27 0.93 to 1.72 1.77 1.21 to 2.58
Cumulative exposure index by quartile,
g/L No. of years†
1 (lowest) 88 112 1.00 1.00
2 101 112 1.05 0.69 to 1.58 1.06 0.62 to 1.82
3 101 112 1.15 0.76 to 1.74 1.23 0.72 to 2.11
4 134 112 1.33 0.89 to 1.99 1.73 1.04 to 2.88
Abbreviations: OR, odds ratio; LD, limit of detection.
The multivariable logistic model includes age (5-year intervals), total plasma lipid concentration (continuous), waist-to-hip ratio ( 0.95, 0.95) and history of
prostate cancer screening (no, yes).
†For subjects with values above LD.
Table 3. Plasma Chlordecone Concentration and Interaction With Family History of Prostate Cancer, and Past Residence in Western Countries
Chlordecone Plasma
Concentration (
g/L)
Without Family History of Prostate
Cancer
With Family History of Prostate Cancer
Interaction
P
Without Past Residence in Western
Countries
With Past Residence in Western
Countries
Interaction
P
No. of
Patients
No. of
Controls OR 95% CI
No. of
Patients
No. of
Controls OR 95% CI
No. of
Patients
No. of
Controls OR 95% CI
No. of
Patients
No. of
Controls OR 95% CI
0.25 (LD) 116 161 1.00 45 26 1.00 137 165 1.00 58 56 1.00
0.25 to 0.47 78 111 1.35 0.80 to 20.26 26 19 0.97 0.33 to 2.83 87 116 1.09 0.68 to 1.74 41 34 1.15 0.53 to 2.48
0.47 to 0.96 81 115 1.13 0.66 to 1.95 34 8 3.22 1.03 to 10.05 103 110 1.12 0.69 to 1.82 36 39 1.33 0.62 to 2.86
0.96 68 123 1.27 0.76 to 2.13 45 14 3.00 1.12 to 8.07 .001 110 118 1.53 0.98 to 2.39 51 31 2.71 1.26 to 5.83 .001
Abbreviations: OR, odds ratio; LD, limit of detection.
The multivariable logistic model includes age (five-year intervals), total plasma lipid concentration (continuous), waist-to-hip ratio ( 0.95, 0.95) and history of
prostate cancer screening (no, yes).
Multigner et al
3460 © 2010 by American Society of Clinical Oncology
J
OURNAL OF CLINICAL ONCOLOGY
Downloaded from ascopubs.org by 37.58.186.129 on February 8, 2017 from 037.058.186.129
Copyright © 2017 American Society of Clinical Oncology. All rights reserved.
increased dramatically during the study period,
15
mostly due to the
development of PSA testing in the population. Consistent with this, it
should be borne in mind that one of the potential negative conse-
quences of prostate cancer screening is an increase in the diagnosis of
indolent or latent tumors that will never be a problem to the patient
and would not have been detected without screening. Such tumors,
like those found in autopsy series,
26
may reflect the natural aging
process rather than the consequences of exposure to chemicals, such as
chlordecone, in the environment.
Cancer is generally thought to occur a long time after the first
exposure and after long periods of continuous exposure. Our study
population fits this requirement, with the median exposure period
being almost 33 years. Moreover, given that chlordecone exposure in
our study population began in adulthood, at a median age of 30.2 years
(the youngest subject being 13 years old at the start of exposure), our
study did not include cases of exposure during critical periods for
carcinogenesis, such as the fetal or neonatal periods.
The prostate cancer risk associated with chlordecone exposure
was higher in subjects with a family history of prostate cancer. Similar
findings were reported for pesticide exposure in the Agricultural
Health Study.
27-30
Study subjects and their first-degree relatives may
have similar patterns of exposure, and this might lead to a statistical
interaction between chlordecone exposure and family history of pros-
tate cancer; alternatively, the observed interaction may be indicative of
an inherited genetic trait, such as a polymorphism in a metabolic
enzyme that alters the balance between bioactivation and detoxifica-
tion in the body.
The prostate cancer risk associated with chlordecone exposure
was particularly marked in subjects who had spent some time living in
a Western country. Most such subjects in our study migrated to a
Western country at the beginning of adulthood, remaining there for a
mean duration of 14 years. Most of these subjects returned to the
French West Indies at the time at which chlordecone use began. Many
intervening factors may explain the interaction observed and any
potential causal relationship should be interpreted with caution. Mi-
gration constitutes a period of exposure to specific environmental risk
factors, including hazardous chemicals or nutritional agents. Residing
in Western countries may induce significant changes in an individual,
due, for example, to the adoption of a Western lifestyle, including, in
particular, eating habits that may be risk factors for prostate cancer.
31
Such changes in lifestyle may then be maintained by these individuals
after their return to the French West Indies.
Previous studies have shown differences in the interindividual
activity of chlordecone reductase in the liver between white and Japa-
nese individuals.
16,32
Such variability has been associated with a pro-
tein variant that arises from two nucleotide (C to G) substitutions at
positions 434 and 931 of the cDNA.
32
We show here that these two
mutations are in complete linkage disequilibrium and that the fre-
quency of the variant allele (0.04) is lower than that previously re-
ported in whites (0.10).
33
Although limited by the small number of
subjects, the observed high risk associated with carrying at least one
variant allele, for individuals with chlordecone concentrations above
the LD, is consistent with the mutated enzyme being less efficient. This
may lead to lower levels of biliary excretion and of chlordecone clear-
ance from the circulation.
Chlordecone may act as a tumor promoter, through hormone-
mediated effects. Chlordecone binds the estrogen receptors
(ER
)
and
(ER
), acting as an agonist of ER
and an antagonist of
ER
.
6,34,35
ER
mediates the adverse effects of estrogen on the pros-
tate, such as aberrant proliferation, inflammation, and malignancy.
ER
exerts opposite and beneficial effects, such as antiproliferative,
anti-inflammatory, and, potentially, anticarcinogenic effects.
36
The
human prostate expresses both ER
and ER
, with ER
expressed
primarily on stromal cells and ER
found in the differentiated epithe-
lium.
37,38
The interplay between the agonistic effects on ER
and the
antagonistic effects on ER
of chlordecone may increase proliferation
of estrogen-sensitive tissues, increasing the risk of cancer. In addition
to its interaction with nuclear ER, chlordecone may activate alterna-
tive estrogen signaling pathways or other enzymes and receptors in-
volved in steroidal homeostasis.
39-43
It remains unclear whether such
mechanisms actually take place in human prostate and whether they
trigger prostate cancer.
We are aware of the inherent limitations of patient-control
studies. Several factors potentially generating bias must be consid-
ered, particularly those relating to differential errors in the mea-
surement of disease or exposure. Patient identification was based
on unequivocal histologic criteria and controls were selected on the
basis of strict criteria, such as normal findings on digital rectal
examination and PSA in the normal range for age, taking into
account the ethnic origin of the population. We recruited incident
rather than prevalent patients, and controls were selected from a
representative sample of the male Guadeloupean population dur-
ing the study period. Exposure was evaluated on the basis of objec-
tive determinations of plasma chlordecone concentration, rather
than on questionnaires, which might generate recall bias. Single
determinations of plasma chlordecone concentration have been
shown to provide an accurate reflection of the load of this com-
pound in the body.
44,45
Classical organochlorine compounds (de-
termined in blood or fat tissues) have been found at lower
concentrations in French West Indies populations than in most
other populations and are not correlated with chlordecone levels.
13
The quantitative evaluation of chlordecone exposure-response is
therefore unlikely to have been confounded by the presence of
other organochlorine compounds in our study. However, we can-
not exclude the possibility that some unknown confounding fac-
tors remain that may account for the associations observed.
In conclusion, our results suggest that there is a causal relation-
ship between chlordecone exposure and prostate cancer risk. Our
study also suggests that this association may be affected by genetic
background, together with environmental agents related to diet or
lifestyle. These findings require further investigation.
AUTHORS’ DISCLOSURES OF POTENTIAL CONFLICTS
OF INTEREST
The author(s) indicated no potential conflicts of interest.
AUTHOR CONTRIBUTIONS
Conception and design: Luc Multigner, Pascal Blanchet
Financial support: Luc Multigner, Marc Romana, Bernard Je´gou,
Pascal Blanchet
Provision of study materials or patients: Helene Delacroix-Maillard,
Pascal Blanchet
Chlordecone Exposure and Prostate Cancer
www.jco.org © 2010 by American Society of Clinical Oncology 3461
Downloaded from ascopubs.org by 37.58.186.129 on February 8, 2017 from 037.058.186.129
Copyright © 2017 American Society of Clinical Oncology. All rights reserved.
Collection and assembly of data: Luc Multigner, Jean Rodrigue Ndong,
Arnaud Giusti, Marc Romana, Helene Delacroix-Maillard,
Pascal Blanchet
Data analysis and interpretation: Luc Multigner, Jean Rodrigue Ndong,
Arnaud Giusti, Marc Romana, Sylvaine Cordier, Bernard Je´gou, Jean
Pierre Thome, Pascal Blanchet
Manuscript writing: Luc Multigner, Jean Rodrigue Ndong, Arnaud
Giusti, Marc Romana, Helene Delacroix-Maillard, Sylvaine Cordier,
Bernard Je´gou, Jean Pierre Thome, Pascal Blanchet
Final approval of manuscript: Luc Multigner, Jean Rodrigue Ndong,
Arnaud Giusti, Marc Romana, Helene Delacroix-Maillard, Sylvaine
Cordier, Bernard Je´gou, Jean Pierre Thome, Pascal Blanchet
REFERENCES
1. Hsing AW, Chokkalingam AP: Prostate cancer
epidemiology. Front Biosci 11:1388-1413, 2006
2. Damber JE, Aus G: Prostate cancer. Lancet
371:1710-1721, 2008
3. Diamanti-Kandarakis E, Bourguignon JP,
Giudice LC, et al: Endocrine-disrupting chemicals:
An Endocrine Society scientific statement. Endocr
Rev 30:293-342, 2009
4. Prins GS: Endocrine disruptors and prostate
cancer risk: Endocr Relat Cancer 15:649-656, 2008
5. Hammond B, Katzzenellenbogen, Krauthammer
N, et al: Estrogenic activity of the insecticide chlorde-
cone (Kepone) and interaction with uterine estrogen
receptors. Proc Natl Acad SciUSA76:6641-6659,
1979
6. Eroschenko VP: Estrogenic activity of the
insecticide chlordecone in the reproductive tract of
birds and mammals. J Toxicol Environ Health 8:731-
742, 1981
7. Snegaroff J: Organochlorinated insecticide
residues in the grounds and the rivers of the ba-
naniere area of Guadeloupe [in French]. Phytiatrie-
Phytopharmacie 26:251-268, 1977
8. Cavelier N: Contamination of fauna by the
organochlorinated pesticides, In Kermarrec A (Ed) :
Current level of the biological chains in Guadeloupe:
Pesticides and heavy metals 1979-1980, INRA,
French Ministry of the Environment [in French], pp
113-128, 1980
9. Cabidoche YM, Achard R, Cattan P, et al:
Long-term pollution by chlordecone of tropical vol-
canic soils in the French West Indies: A simple
leaching model accounts for current residue. Envi-
ron Pollut 157:1697-1705, 2009
10. Bocquene´ G, Franco A: Pesticide contamina-
tion of the coastline of Martinique. Mar Pollut Bull
51:612-619, 2005
11. Dubuisson C, He´ raud F, Leblanc JC, et al:
Impact of subsistence production on the manage-
ment options to reduce the food exposure of the
Martinican population to Chlordecone. Regul Toxicol
Pharmacol 49:5-16, 2007
12. Multigner L, Kadhel P, Huc-Terki F, et al:
Exposure to chlordecone and male fertility in Guade-
loupe (French West Indies). Epidemiology 17: 6,
2006,(suppl; abstr S372)
13. Kadhel P: Pesticides in the Antilles, impact on
the function of reproduction [in French]. PhD thesis.
Universite´ des Antilles et de la Guyane, Guadeloupe,
French West Indies, 2008
14. Sirica AE, Wilkerson CS, Wu LL, et al: Evalu-
ation of chlordecone in a two-stage model of hepa-
tocarcinogenesis: A significant sex difference in the
hepatocellular carcinoma incidence. Carcinogenesis
10:1047-1054, 1989
15. Mallick S, Blanchet P, Multigner L: Prostate
cancer incidence in Guadeloupe, a French Caribbean
Archipelago. Eur Urol 47:769-772, 2005
16. Molowa DT, Shayne AG, Guzelian PS: Purifica-
tion and characterization of chlordecone reductase
from human liver. J Biol Chem 261:12624-12627,
1986
17. Cussenot O, Azzouzi AR, Nicolaiew N, et al:
Combination of polymorphisms from genes related
to estrogen metabolism and risk of prostate can-
cers: The hidden face of estrogens. J Clin Oncol
25:3596-3602, 2007
18. Ritchie JM, Vial SL, Fuortes LJ, et al: Compari-
son of proposed frameworks for grouping polychlori-
nated biphenyl congener data applied to a case-control
pilot study of prostate cancer. Environ Res 98:104-
113, 2005
19. Hardell L, Andersson SO, Carlberg M, et al:
Adipose tissue concentrations of persistent organic
pollutants and the risk of prostate cancer. J Occup
Environ Med 48:700-707, 2006
20. Aronson KJ, Wilson JW, Hamel M, et al:
Plasma organochlorine levels and prostate cancer
risk. J Expo Sci Environ Epidemiol 10.1038/
jes.2009.33 [epub ahead of print on June 10, 2009]
21. Xu X, Dailey AB, Talbott EO, et al: Associa-
tions of Serum Concentrations of Organochlorine
(OC) pesticides with breast cancer and prostate
cancer in U.S. adults. Env Health Perspect 118:60-
66, 2010
22. Hsing AW, Deng J, Sesterhenn IA, et al: Body
size and prostate cancer: A population-based case-
control study in China. Cancer Epidemiol Biomarkers
Prev 9:1335-1341, 2000
23. Jackson MD, Walker SP, Simpson CM, et al:
Body size and risk of prostate cancer in Jamaican
men. Cancer Causes Control 21:909-917, 2010
24. Agudo A, Gon˜ i F, Etxeandia A, et al: Polychlo-
rinated biphenyls in Spanish adults: Determinants of
serum concentrations. Environ Res 109:620-628,
2009
25. Weiss NS: Adjusting for screening history in
epidemiologic studies of cancer: Why, when, and
how to do it. Am J Epidemiol 157:957-961, 2003
26. Adolfsson J: Commentary: On the incidence
of histological prostate cancer and the probable
diagnosis of cases with tumors too small to produce
symptoms or to attract attention on physical exam-
ination: The findings of Dr Arnold Rice Rich. Int J
Epidemiol 36:285-287, 2007
27. Alavanja MC, Samanic C, Dosemeci M, et al:
Use of agricultural pesticides and prostate cancer
risk in the Agricultural Health Study cohort. Am J
Epidemiol 157:800-814, 2003
28. Mahajan R, Bonner MR, Hoppin JA, et al:
Phorate exposure and incidence of cancer in the
Agricultural Health Study. Environ Health Perspect
114:1205-1209, 2006
29. Lynch SM, Mahajan R, Beane Freeman LE, et
al: Cancer incidence among pesticide applicators
exposed to butylate in the Agricultural Health Study
(AHS). Environ Res 109:860-868, 2009
30. Christensen CH, Platz EA, Andreotti G, et al:
Coumaphos exposure and incident cancer among
male participants in the Agricultural Health Study
(AHS). Environ Health Perspect 118:92-96, 2010
31. Chan JM, Gann PH, Giovannucci EL: Role of
diet in prostate cancer development and progres-
sion. J Clin Oncol 23:8152-8160, 2005
32. Kume T, Iwasa H, Shiraishi H, et al: Character-
ization of a novel variant (S145C/L311V) of 3alpha-
hydroxysteroid/dihydrodiol dehydrogenase in human
liver. Pharmacogenetics 9:763-771, 1999
33. National Center for Biotechnology Informa-
tion. http://www.ncbi.nlm.nih.gov/SNP/
34. Kuiper GG, Lemmen JG, Carlsson B, et al:
Interaction of estrogenic chemicals and phytoestro-
gens with estrogen receptor beta. Endocrinology
139:4252-4263, 1998
35. Lemaire G, Mnif W, Mauvais P, et al: Activa-
tion of alpha- and beta-estrogen receptors by persis-
tent pesticides in reporter cell lines. Life Sci 79:
1160-1169, 2006
36. Ellem SJ, Risbridger GP: The dual, opposing
roles of estrogen in the prostate. Ann N Y Acad Sci
1155:174-186, 2009
37. Schulze H, Claus S: Histological localization of
estrogen receptors in normal and diseased human
prostates by immunocytochemistry. Prostate 16:
331-343, 1990
38. Enmark E, Pelto-Huikko M, Grandien K, et al:
Human estrogen receptor
-gene structure, chro-
mosomal localization, and expression pattern. J Clin
Endocrinol Metabol 82:4258-4265, 1997
39. Thomas P, Dong J: Binding and activation
of the seven-transmembrane estrogen receptor
GPR30 by environmental estrogens: A potential
novel mechanism of endocrine disruption. J Steroid
Biochem Mol Biol 102:175-179, 2006
40. Ray S, Xu F, Li P, et al: Increased level of
cellular bip critically determines estrogenic potency
for a xenoestrogen kepone in the mouse uterus.
Endocrinology 148:4774-4785, 2007
41. Scippo ML, Argiris C, Van De Weerdt C, et al:
Recombinant human estrogen, androgen and pro-
gesterone receptors for detection of potential endo-
crine disruptors. Anal Bioanal Chem 378:664-669,
2004
42. Benachour N, Moslemi S, Sipahutar H, et al:
Cytotoxic effects and aromatase inhibition by xeno-
biotic endocrine disrupters alone and in combina-
tion. Toxicol Appl Pharmacol 222:129-140, 2007
43. Lee J, Scheri RC, Zhang Y, et al: Chlordecone,
a mixed pregnane X receptor (PXR) and estrogen
reeptor alpha (ERalpha) agonist, alters cholesterol
homeostasis and lipoprotein metabolism in C57BL/6
mice. Toxicol Appl Pharmacol 233:193-202, 2008
44. Cannon SB, Veazey KM, Jackson RS, et al:
Epidemic Kepone poisoning in chemical workers.
Am J Epidemiol 107:529-537, 1978
45. Cohn WJ, Boylan JJ, Blanke RV, et al: Treat-
ment of chlordecone (Kepone) toxicity with cho-
lestyramine resultas of a controlled trial. N Engl
J Med 298:243-248, 1979
■■■
Multigner et al
3462 © 2010 by American Society of Clinical Oncology
J
OURNAL OF CLINICAL ONCOLOGY
Downloaded from ascopubs.org by 37.58.186.129 on February 8, 2017 from 037.058.186.129
Copyright © 2017 American Society of Clinical Oncology. All rights reserved.