You are on page 1of 10

Cancer Causes and Control 14: 805–814, 2003.

805
 2003 Kluwer Academic Publishers. Printed in the Netherlands.

Smoking and cervical cancer: pooled analysis of the IARC multi-centric case–control
study

Martyn Plummer1,*, Rolando Herrero2, Silvia Franceschi1, Chris J.L.M. Meijer3, Peter Snijders3, F. Xavier Bosch4,
Silvia de Sanjosé4 & Nubia Muñoz1 for the IARC Multi-centre Cervical Cancer Study Group 
1
International Agency for Research on Cancer, 150 Cours Albert Thomas, F-69372 Lyon, France; 2Proyecto
Epidemiologic Guanacaste, Costa Rica; 3Department of Pathology, VU Medical Center, Amsterdam, The
Netherlands; 4Epidemiology and Cancer Registry Service, Catalan Institute of Oncology (ICO), Barcelona, Spain

Received 24 February 2003; accepted in revised form 24 June 2003

Key words: Cervical cancer, human papillomavirus, smoking.

Abstract

Background: Smoking has long been suspected to be a risk factor for cervical cancer. However, not all previous
studies have properly controlled for the effect of human papillomavirus (HPV) infection, which has now been
established as a virtually necessary cause of cervical cancer. To evaluate the role of smoking as a cofactor of
progression from HPV infection to cancer, we performed a pooled analysis of 10 previously published case–control
studies. This analysis is part of a series of analyses of cofactors of HPV in the aetiology of cervical cancer.
Methods: Data were pooled from eight case–control studies of invasive cervical carcinoma (ICC) and two of
carcinoma in situ (CIS) from four continents. All studies used a similar protocol and questionnaires and included a
PCR-based evaluation of HPV DNA in cytological smears or biopsy specimens. Only subjects positive for HPV
DNA were included in the analysis. A total of 1463 squamous cell ICC cases were analyzed, along with 211 CIS
cases, 124 adeno- or adeno-squamous ICC cases and 254 control women. Pooled odds ratios (OR) and 95%
confidence intervals (CI) were estimated using logistic regression models controlling for sexual and non-sexual
confounding factors.
Results: There was an excess risk for ever smoking among HPV positive women (OR 2.17 95%CI 1.46–3.22). When
results were analyzed by histological type, an excess risk was observed among cases of squamous cell carcinoma for
current smokers (OR 2.30, 95%CI 1.31–4.04) and ex-smokers (OR 1.80, 95%CI 0.95–3.44). No clear pattern of
association with risk was detected for adenocarcinomas, although the number of cases with this histologic type was
limited.
Conclusions: Smoking increases the risk of cervical cancer among HPV positive women. The results of our study are
consistent with the few previously conducted studies of smoking and cervical cancer that have adequately controlled
for HPV infection. Recent increasing trends of smoking among young women could have a serious impact on
cervical cancer incidence in the coming years.

Introduction cause of cervical cancer [1, 2]. This discovery has


important implications for both primary and secondary
Infection with certain types of human papillomavirus prevention, with the prospect that the disease may
(HPV) has been established as a virtually necessary eventually be eliminated by prophylactic vaccination.
Current knowledge of the aetiology of cervical cancer,
* Address correspondence to: Martyn Plummer, Unit of Field and however, remains incomplete. Cervical cancer is a rare
Intervention Studies, International Agency for Research on Cancer,
consequence of HPV infection and, when it does occur,
150 Cours Albert Thomas, F-69372 Lyon, France. Ph.: +33-472-
738446/738402; Fax: +33-472-738345; E-mail: plummer@iarc.fr it develops many years after initial infection. For these
 
See Appendix 1 for details. reasons, current research efforts are oriented towards
806 M. Plummer et al.
identifying cofactors that may influence the progression cell carcinoma, adenocarcinoma and adenosquamous
from infection to cancer. Cofactors may be character- carcinoma. The last two categories have been combined
istics of the virus or host, or may be environmental. for this analysis.
Between 1985 and 1997 the International Agency for For all studies except the two ICC studies in Spain
Research on Cancer conducted a series of case–control and Colombia, controls were hospital based, and were
studies of cervical cancer; eight studies on invasive frequency matched by age in five-year age groups.
cancer [3–10] and two on carcinoma in situ (CIS) [11, Diseases associated with known risk factors for cervical
12]. All studies used a similar protocol and question- neoplasia, including tobacco consumption, were exclud-
naires and included an accurate evaluation of HPV ed. For the two CIS studies, the controls were popula-
DNA in cytological smears or biopsy specimens. This tion based and were individually matched by age and
report is part of a series of papers [13–16] that use the date of cytology. The matching has been ignored in the
pooled data from these studies to evaluate possible current analysis, with no material effect on the results.
cofactors for cervical cancer. We currently believe that All consenting participants were administered a stan-
the appropriate way to evaluate potential cofactors, dardized questionnaire including information on socio-
while controlling for the effect of HPV, is to restrict the demographic factors, sexual behaviour, reproductive
analysis to women with HPV infections. history, contraceptive practices, smoking habits, genital
Smoking has long been associated with cervical cancer hygiene, history of STDs and history of cervical
risk [17]. However, it has been difficult to rule out cytological screening. Face-to-face interviews were con-
residual confounding, chiefly from sexual habits, as an ducted in hospital by trained interviewers. In the part of
explanation for this association [18, 19]. In particular, the questionnaire on tobacco, subjects were first asked
few studies of smoking and cervical cancer have to classify themselves as life long non-smokers, current
controlled for HPV infection. This study allows us to smokers or ex-smokers (defined as a former smoker who
examine the role of smoking while controlling for the stopped smoking at least 1 year prior to the interview).
strong effect of HPV infection and, additionally, other Ever-smokers were then asked the age at which they
cofactors that have been identified in the same popula- started smoking, how many cigarettes per day they
tions [13, 14]. smoked and the total time made up of periods (if any) in
which they stopped smoking. Ex-smokers were asked
the age at which they finally stopped smoking. Further
Subjects and methods information on duration and number of cigarettes was
collected by type of tobacco. Information on duration
This analysis concerns 1798 HPV positive cases and 254 and number of times per day was also collected for
HPV positive controls from eight separate case–control certain chewing habits: chewing tobacco, taking snuff
studies of invasive cervical cancer (ICC) and two studies and chewing betel quid (with and without tobacco).
of CIS. The ICC studies were conducted in eight After the interview, all subjects had a pelvic examination
countries, which showed wide variation in incidence performed by a gynaecologist, in which exfoliated cells
rates. Regions covered include Thailand [5] and the for cytological examination and HPV analysis were
Philippines [8] in Asia; Morocco [4] in Africa; Brazil taken. A tumour biopsy specimen was also taken in
[6], Peru [10], Paraguay [9] and Colombia [3, 7] in South most cases and kept frozen.
America and Spain [3, 7] in Europe. The two CIS
studies were performed in Spain and Colombia at HPV DNA detection
the same time as the studies of ICC [11, 12]. These have
also been included in this analysis because CIS is Exfoliated cells were collected using wooden spatulae
accepted as an immediate precursor of ICC, and and endocervical brushes. Two scrapes were obtained
because it shared the same risk factors in our studies from each woman. After preparation of one Pap smear,
[20]. the remaining cells were eluted in phosphate-buffered
Methods and findings from each study have been saline (PBS), peleted in PBS and kept frozen at )70 C
previously described. In summary, eligible patients were until shipment to a central laboratory for HPV testing.
histologically confirmed, incident ICC or CIS identified Detailed descriptions of the hybridization assays
at the reference hospitals. Cases had no previous employed are given in the individual publications of
treatment for the disease and gave informed consent to the various studies. Briefly, HPV DNA detection and
participate in the study. The histological diagnosis was typing were performed in central laboratories using
reviewed in each study either by a single expert, or by a PCR amplification methods targeting a small fragment
panel. Cases were histologically classified as squamous of the L1 gene. DNA quality was evaluated using
Pooled analysis of smoking and cervical cancer 807
b-globin primers. In the studies from Colombia and reported in only a few centres. These exposures have
Spain, MY09 and MY11 consensus primers were used been combined into a single variable for ‘chewing’ which
[21]. In the remaining studies GP5+/6+ primers were was controlled for in the final analysis. Other confound-
employed. PCR products were assessed for HPV posi- ers considered and their categories were age (in five year
tivity by low-stringency Southern blot hybridization bands), centre, education (none, primary, secondary+),
using a cocktail of HPV-specific probes [22]. For viral number of sexual partners (0–1, 2, 3, 4+), age at first
genotyping the PCR products were successively hybrid- intercourse (under 16, 17–21, 22+), years of oral
ized with oligonucleotide probes for 33 HPV types as contraceptive use (never, 1–4, 5+ years), parity (0, 1–
previously described [22, 23]. HPV DNA-positive spec- 2, 3+) and screening history (never, ever, excluding
imens that did not hybridize with any of the 33 probes screening visits 12 months before the interview).
were tentatively called HPV X.
In a second step, E7 primers for 14 high-risk HPV
types [2] were used for reamplification of the following Results
specimens: all case specimens positive for the b-globin
gene and classified as HPV X or HPV DNA negative, all Table 1 shows, for each study area, data on incidence
specimens from control women classified as HPV X, and rates of cervical cancer, prevalence of HPV infection in
a subsample of specimens from control women found to cases and controls, and prevalence of tobacco consump-
be b-globin-positive and HPV DNA-negative. Standard tion. Incidence rates are taken from Globocan 2000 [25].
precautions were taken to minimize the risk of false- They show that Spain is a low-risk country, Morocco,
positive results in the PCR reactions as described Thailand and the Philippines are intermediate risk
elsewhere [24]. All PCR assays were performed blind countries, and the South American countries are high
to the case–control status. Failure to detect HPV DNA risk. As expected, prevalence of HPV infection is very
with a negative amplification of b-globin led to an high in cases, but low in controls, ranging from 6% in
invalid result and these women have been excluded from Spain to 21% in Morocco. Smoking prevalence in
analysis. controls is low in most countries, with the notable
Because some of the re-testing was performed at the exceptions of Brazil (36% ever smokers), Colombia
completion of the fieldwork in all study areas, numbers (28% ever smokers), and the CIS study in Spain (38%
of HPV positive cases and controls of this analysis ever smokers). The average number of cigarettes per day
sometimes differ from those reported in the original among smokers is also low (3–11 cigarettes per day in
publications. controls). Chewing is relatively common in only two
centres: Peru where 5% of controls were chewers,
mainly of coca leaves, and Thailand where 20% of
Statistical analysis controls were chewers, maily of betel.
Figure 1 summarizes the OR of cervical cancer for
Logistic regression was used to estimate odds ratios ever-smokers compared with never-smokers in each
(ORs) and 95% confidence intervals (95%CI). Likeli- study and the pooled results. ORs in this figure were
hood ratio tests were used to assess the significance of controlled only for age and centre. Ever smoking is
terms in the model. Heterogeneity of the OR between positively associated with cervical cancer (OR ¼ 2.17,
centres, and other stratifying variables, was tested by 95%CI 1.46–3.22). The results for each centre are
introducing an interaction term between smoking and relatively homogeneous: the formal test for heterogene-
the modifying factor. ity has a p-value of 0.11. However, Morocco is a clear
Most results are presented graphically, plotting the outlier which shows a negative association (OR ¼ 0.27,
summary ORs as a black square, whose size is inversely 95%CI 0.05–1.45). This may be due to a design flaw.
proportional to the variance of the estimate. A hori- Cases in the Moroccan study were drawn from a
zontal line represents the 95%CI. Diamonds are used to specialist cancer hospital whereas controls were recruit-
plot the summary OR for squamous cell ICC, CIS and ed from a general hospital with a smaller catchment
all studies together. The centre of the diamond repre- area. In the original analysis of this data, an attempt was
sents the pooled OR and the extremes show the limits of made to overcome this selection bias by controlling for
the 95%CI. A log scale is used for the OR in order to social class and urban/rural residence [4]. The OR in
preserve symmetry of the CI. Morocco controlling for education (as a marker of SES)
This report considers only tobacco consumption in and residence is 0.63 (95%CI 0.08–4.8). Morocco was
the form of cigarette smoking. Chewing of tobacco or retained in the analysis, but with an additional adjust-
other substances (coca, betel) and snuff taking was ment for urban/rural residence.
808 M. Plummer et al.
Table 1. HPV prevalence and smoking habits by centre

ASRa HPV tested HPV positive Ever smokers (%)


(world)
Cases Controls Cases (%) Controls (%) Cases Controls

Invasive cancer
Paraguay 41.1 112 90 109 (97.3) 17 (18.9) 32.2 19.0
Peru 39.9 196 175 186 (94.9) 31 (17.7) 9.6 3.6
Colombia 32.9 110 126 86 (78.2) 22 (17.5) 42.4 28.1
Brazil 31.3 187 196 181 (96.8) 34 (17.3) 47.2 36.0
Philippines 22.7 364 381 349 (95.9) 35 (9.2) 20.4 7.2
Thailand 20.7 378 261 363 (96.0) 41 (15.7) 17.1 13.0
Morocco 18.8 188 176 182 (96.8) 38 (21.6) 2.8 4.4
Spain 7.2 159 136 131 (82.4) 8 (5.9) 20.4 14.3
Carcinoma in situ
Colombia 135 181 96 (71.1) 19 (10.5) 37.2 23.7
Spain 157 193 115 (73.2) 9 (4.7) 54.5 38.4
Total 1986 1915 1798 254

a
Age standardized incidence rate from Globocan 2000 [25].
b
Among ever smokers.

Fig. 1. OR for ever smoking by study.

As a further test to ensure that the results are not was recalculated after leaving out each study in turn.
unduly influenced by any single study, the pooled OR The pooled OR remained fairly stable, ranging from
Pooled analysis of smoking and cervical cancer 809

Fig. 2. ORs of squamous cell carcinoma of the cervix.

1.94 (95%CI 1.29–2.94) when the Philippines were noma and adenosquamous carcinoma were also exam-
removed to 2.37 (95%CI 1.54–3.67) when the ICC ined but since the number of cases was small, the results
study in Colombia was removed. are not presented in detail. There were 82 HPV positive
Figure 2 shows pooled results for various aspects of adenocarcinomas (2.0% of cases) and 42 adenosqua-
smoking history for squamous cell carcinoma (including mous carcinomas (1.0% of cases). When these were
CIS), controlling for age, centre, education, number of combined, the OR for ever smokers was 1.64 (95%CI
sexual partners, age at first intercourse, oral contracep- 0.64–4.21). When adenocarcinomas were examined
tive use, parity and screening. Both current and ex- on their own the OR was 2.19 (95%CI 0.74–6.47).
smokers showed elevated risks. However, among ever The results for these histological types are therefore
smokers, there is no evidence of increasing risk with compatible with no excess risk for smokers, but they are
years of smoking, number of cigarettes per day or age at also compatible with the results for squamous cell
starting smoking. In order to confirm this finding, trend carcinoma.
tests were conducted for these three risk factors, treating Figure 3 shows the modifying effect of age and
them as continuous variables. The resulting p-values number of sexual partners on the OR for ever smoking.
were 0.99 for duration, 0.90 for number of cigarettes per Results are presented for all histological types com-
day and 0.52 for age at starting smoking. Adenocarci- bined. There is no strong evidence of heterogeneity

Fig. 3. ORs for ever smoking stratified by age and number of sexual partners.
810 M. Plummer et al.
Table 2. ORs of HPV-positivity and 95% CI according to reproductive factors among control women

HPV prevalence (%) HPV positive/HPV negative ORa (95%CI)

Smoking status
Never 14 218/1369 1.00
Current 10 22/195 0.85 (0.52, 1.39)
Ex 13 14/97 0.85 (0.47, 1.56)
Test for heterogeneity p = 0.72
No. of years smoking
Never 14 218/1369 1.00
1–19 11 19/158 1.04 (0.61, 1.77)
‡20 11 15/127 0.65 (0.36, 1.16)
Test for trend p = 0.21
No. cigarettes/day
Never 14 218/1369 1.00
<6 12 17/129 0.84 (0.49, 1.45)
‡6 11 18/151 0.92 (0.53, 1.58)
Test for trend p = 0.61
Age started smoking
Never 14 218/1369 1.00
‡20 14 18/117 1.07 (0.63, 1.83)
15–19 10 13/111 0.93 (0.50, 1.72)
<15 8 5/59 0.45 (0.17, 1.18)
Test for trend p = 0.22
Years since quitting
Current smoker 10 22/195 1.00
1–10 15 10/55 1.52 (0.62, 3.74)
‡11 7 3/38 0.60 (0.16, 2.35)
Test for trend p = 0.78

a
Controlling for study centre and age.

across either age groups (p ¼ 0.16 for heterogeneity test) cervical cancer that persists after controlling for the
or number of sexual partners (p ¼ 0.17). strong effect of HPV and for other potential cofactors of
The association with smoking was also tested after progression from infection to cancer. The low level of
further restriction to subjects infected with high risk smoking in most of the study populations hampered our
HPV types (16, 18, 26, 31, 33, 35, 39, 45, 51, 52, 53, 56, ability to study the presence of a dose-response effect.
58, 59, 66, 68, 73, 82). This subgroup comprised 93% of The hypothesis that smoking is a risk factor for
the HPV positive cases (1667 cases) and 63% of the cervical cancer was first put forward in 1977 by
HPV positive controls (161 controls). The association Winkelstein [17], who also periodically reviewed the
with ever smoking was not changed (OR ¼ 2.14, 95%CI support for this hypothesis from subsequent epidemio-
1.22–3.78, controlling for all confounders listed above). logical studies [26, 27]. Despite a consistent association
There was also no evidence of any dose–response between smoking and cervical cancer after an adjust-
relationship (data not shown). ment for sexual behaviour, it was not generally agreed
In addition to analysing smoking as a risk factor for that confounding could be ruled out as an explanation
cervical cancer among HPV positive women, smoking for this finding, since sexual behaviour was evidently a
was also examined as a risk factor for HPV acquisition surrogate for a sexually transmitted agent [18, 19, 28].
among controls. The results are presented in Table 2 The hypothesis that HPV is the causal agent was first
and show no association between smoking behaviour put forward by zur Hausen [29]. Now that HPV has
and HPV. been identified as the principal cause of cervical cancer,
it should be possible to resolve the controversy over
smoking. However, relatively few studies have con-
Discussion trolled for HPV when examining smoking. Of these,
some have used a relatively inaccurate HPV DNA
Our present pooled analysis of 1798 cases or cervical detection method [30] or anti-HPV antibodies [31], and/
carcinoma and 254 control women who were infected or have concentrated on a limited range of HPV types
with HPV shows that ever-smokers have an excess risk of [30–33].
Pooled analysis of smoking and cervical cancer 811
The evidence for pre-invasive cervical neoplasias is Various possible mechanisms have been proposed for
larger than for invasive ones. In a clinic-based case– the association between smoking and cervical cancer. In
control study of high grade dysplasia in the USA, Becker animal models, more than 50 years ago Rous and
et al. [34] found an increased risk for current smokers Friedwald [41] described the malignant conversion of
that persisted after adjusting for HPV, age at first sexual cottontail rabbit papillomavirus after exposure to me-
intercourse, number of sexual partners and ethnicity thilcolanthrene or tar. More recently, malignant trans-
(OR ¼ 1.8, 95%CI 1.2–2.8). No excess risk was observed formation has been observed when endocervical cells
for ex-smokers. Kjellberg et al. [35] conducted a popu- immortalized by HPV 16 are exposed to cigarette smoke
lation-based study of high-grade cervical intraepithelial condensate, whereas untreated cells remain non-tumori-
neoplasia (CIN) in Sweden. An excess risk of CIN 2–3 genic in nude mice [42].
was observed for ever-smoking that persisted after Tobacco is able to induce its carcinogenic effect in
adjusting for HPV DNA positivity (OR 2.5, 95%CI sites not directly exposed to cigarette smoke, as evi-
1.3–4.9). Deacon et al. [36] conducted a nested case– denced by associations of smoking with pancreatic,
control study of CIN 3 in England. In contrast to the kidney and bladder cancer [43]. In the cervix, it is
previously reported studies, which used multiple regres- possible to detect nicotine derivatives like cotinine and
sion to control for the effect of HPV, Deacon et al. tobacco specific nitrosamines (recently reviewed by
separately analyzed risk factors for progression among Szarewski and Cuzick [44]). In addition, DNA adducts
HPV positive women and risk factors for presence of [45] and other evidence of genotoxic damage [46] are
HPV among control women. Ever smoking was a risk detectable in cervical exfoliated cells. It has been shown
factor for CIN 3 among HPV positive women (OR ¼ 2.2, that smoking affects local immune mechanisms in the
95%CI 1.44–3.35). The excess risk persisted after adjust- cervix, with reductions in the number of Langerhans
ing for age at first intercourse, number of sexual partners, cells and other markers of immune function [47].
spontaneous abortion and time since start of latest Another possibility is a systemic effect of smoking, by
regular relationship. There was also strong evidence of a which smokers would have an altered metabolism of
dose–response effect. By contrast, smoking was not female hormones compared with non-smokers. The
associated with HPV DNA positivity among controls association presented here with smoking remains after
(OR ¼ 0.90, 95%CI 0.60–1.35). Hildesheim et al. [37] controlling for oral contraceptive use and parity, but we
conducted a population based study in Costa Rica using do not have sufficient data to analyze the interactions
prevalent cases of HSIL or cancer as an outcome. Both between these exposures.
current smokers and ex-smokers were at increased risk, Our approach of restricting the analysis to HPV
with ORs of 2.4 (95%CI 1.2–5.1) and 2.3 (95%CI 1.3– positive women is based on our current understanding
4.3) respectively, after controlling for age, HPV type of HPV as a virtually necessary cause of cervical cancer.
(high-risk versus low-risk) and parity. Finally, Lacey This implies that if smoking is a risk factor for cervical
et al. [38] conducted a multi-centre case–control study of cancer, it may act either by increasing the risk of
cervical adenocarcinoma in the USA, using community acquiring an HPV infection, or by increasing the risk of
controls and, additionally an age-matched group of SCC progression from HPV infection to cancer. We found no
cases [38]. After controlling for HPV, education, number association between smoking and HPV infection among
of sexual partners and screening history, ever smoking controls. Conversely, we found an increased risk of cer-
was not associated with adenocarcinoma (OR ¼ 0.8, vical cancer for smokers among HPV positive women.
95%CI 0.5–1.2) or SCC (OR ¼ 1.4, 95%CI 0.8–2.3). Hence we conclude that smoking acts as a risk factor for
There was limited evidence that heavy smoking was progression once HPV infection is acquired. This
associated with SCC (OR ¼ 1.8, 95%CI 1.0–3.3 for restriction should be borne in mind when interpreting
‡1 pack/day) but not with adenocarcinoma (OR ¼ 0.7, the results: the ORs presented here cannot be extrapo-
95%CI 0.4–1.3 for ‡1 pack/day). The results of the lated to the general population, but are restricted to
present analysis are therefore broadly consistent with HPV positive women. In order to check our conclusions
other case–control studies that have controlled for HPV against a more traditional analysis, we re-examined the
infection using high quality HPV assays. In one of the relationship between ever smoking and risk of cervical
few available prospective studies, tobacco smoke was cancer by multiple logistic regression using the full set of
associated with progression from CIN I to CIN III subjects on whom an HPV test result was available
among HPV 16 positive women [39]. Significantly higher (1986 cases and 1915 controls). The OR for ever-
regression rates of cervical precancerous lesions have also smoking controlling for HPV, age and centre was 2.32
been reported for women who quit smoking compared to (95%CI 1.78–3.04). Further control for education,
those who continue with the habit [40]. screening, parity, number of sexual partners, oral
812 M. Plummer et al.
contraceptive use and age at first intercourse did not adequately controlled for HPV infection when examin-
substantially alter this result (OR 2.31, 95%CI 1.72– ing the association between smoking and cervical
3.11). cancer. Taken together, this body of evidence suggests
One of the limitations of restricting the analysis to that squamous cell carcinoma of the cervix should be
HPV positive women in case control studies is that only added to the list of tobacco associated cancers, while for
the current HPV status is available. Ideally the analysis adenocarcinoma, further data are warranted. This
should be restricted to women with chronic infections conclusion was supported by an expert working group
since this is the underlying risk factor. Whereas all cases convened by the International Agency for Research on
will necessarily have chronic HPV infections, this is not Cancer in June 2002 [48].
necessarily true for all HPV positive controls. This Several countries are experiencing an increase in the
means that control for chronic HPV infection might be prevalence of smoking, particularly among younger
incomplete. We have examined this possibility by testing women [49]. This increase in smoking among women
the modifying effect of age and number of sexual could have a serious impact on the incidence of cervical
partners – two risk factors that may be associated with cancer in the future, emphasizing the need for intensi-
chronic infection. The null results of this test suggest fying preventive efforts [50].
that chronic infection has been adequately controlled for
in the statistical model.
Apart from HPV infection, we have tried to control
Appendix 1
for three separate causes of confounding: screening,
sexual behaviour and reproductive factors. Screening The group is composed of the following researchers:
has been observed to be protective against ICC in most S. Franceschi, M. Plummer, J. Smith, N. Muñoz, IARC, Lyon,
of these populations. Sexual behaviour is a surrogate for France; F.X. Bosch, V. Moreno, S. de Sanjosé, X. Castellsagué X,
other sexually transmitted infections that may accelerate ICO, Barcelona, Spain; R. Herrero, Proyecto Epidemiologico Gua-
the progression to cancer. Chlamydia and Herpes nacaste, Costa Rica; C.J.L.M. Meijer, J.M.M. Walboomers and P.J.F.
Snijders, Free University Hospital, Amsterdam, the Netherlands; K.V.
Simplex virus 2 are two such infectious agents that will Shah, Johns Hopkins University, Baltimore, Maryland; S. Chichareon,
be investigated more closely in future papers. Parity and Prince of Songkla University, Hat-Yai, Thailand; C. Ngelangel,
oral contraceptive have been previously identified in University of The Philippines, Manila, the Philippines; N. Chaoki,
these studies as hormonal cofactors of progression in B. El Gueddari, Institut National d’Oncologie, Rabat, Morocco;
this pooled data [13, 14]. J. Eluf-Neto, Universidade de Sao Paulo, Sao Paulo, Brazil; P.A.
Rolón, Laboratorio de Anatomia Patológica y Citologia, Asunción,
Smoking is rare among women in developing coun- Paraguay; C. Santos, E. Cacares, Maes Heller Cancer Research
tries and in Spain. Ideally, the relationship between Institute, Lima, Peru; S. Bayo, Institut National de Recherche en Santé
smoking and cervical cancer should be addressed in Publique, Bamako, Mali; I. Izarzugaza, Euskadi Cancer Registry,
populations in which smoking is more common among Vitoria Gasteiz, Spain; M. Gili, Catedra Reynals, Barcelona, Spain;
women. However, opportunities to study large numbers L.A. Tafur, University of Valle, Cali, Colombia; C. Navarro, Health
Council, Murcia, Spain; N. Ascunce, Breast Cancer Prevention Centre,
of women with invasive cervical cancer are rare, and we Pamplona, Spain; L.C. González, Delegation of Social Welfare,
have taken advantage of the possibilities offered by this Salamanca, Spain; M. Santamaria, Navarra Hospital, Pamplona,
series of studies with a common protocol and accurate Spain; P. Alonso de Ruiz, General Hospital of Mexico, Mexico City,
HPV measurements. Mexico; N. Aristizabal, Cali, Colombia; J. Deacon, Institute of Cancer
Selection bias is always a potential problem in case– Research, Belmont, United Kingdom.
control studies. This issue has been discussed in the
original reports for these studies, with the generally
reassuring conclusions that there is no problem. The Acknowledgements
only exception is Morocco, in which we have attempted
to overcome the evident selection bias by controlling for This research was funded by the European Community
urban/rural residence. As smoking is associated with a CI 1-0371-F(CD); The Fondo de Investigaciones Sani-
broad range of diseases, hospital controls may include tarias (FIS), Spain 86/753, 87/1513, 88/2049, 90/0901,
an excess of smokers compared to the source popula- 95/0955; Preventiefonds, The Netherlands 28-1502.1;
tion. However, in our studies patients in hospital for Programa Interministerial de Investigación y Desarrol-
smoking-related diseases were not eligible as control lo, Spain SAF 96/0323, The Conselho Nacional de
women. Desenvolvimiento Cientifico e Tecnologico, Brazil
In conclusion, smoking increases the risk of cervical (CNPq) JEN-204453/88.7 and the Department of Re-
cancer among HPV positive women. The present pooled productive Health & Research at the World Health
analysis adds to the limited number of studies that have Organization, grant no. 98.101.
Pooled analysis of smoking and cervical cancer 813
References M, eds. Molecular Diagnostics of Human Cancer. New York: Cold
Spring Harbor Press, pp. 209–214.
1. IARC (1995) IARC Monographs on the Evaluation of Carcinogenic 22. Jacobs MV, de Roda Husman AM, van den Brule AJ, Snijders PJ,
Risks to Humans, volume 64. Human Papillomaviruses. Lyon: Meijer CJ, Walboomers JM (1995) Group-specific differentiation
International Agency for Research on Cancer. between high- and low-risk human papillomavirus genotypes by
2. Walboomers JM, Jacobs MV, Manos MM, et al. (1999) Human general primer-mediated PCR and two cocktails of oligonucleotide
papillomavirus is a necessary cause of invasive cervical cancer probes. J Clin Microbiol 33: 901–905.
worldwide. J Pathol 189: 12–19. 23. Roda Husman AM, Walboomers JM, Meijer CJ, et al. (1994)
3. Bosch FX, Munoz N, de Sanjose S, et al. (1992) Risk factors for Analysis of cytomorphologically abnormal cervical scrapes for the
cervical cancer in Colombia and Spain. Int J Cancer 52: 750–758. presence of 27 mucosotropic human papillomavirus genotypes,
4. Chaouki N, Bosch FX, Munoz N, et al. (1998) The viral origin of using polymerase chain reaction. Int J Cancer 56: 802–806.
cervical cancer in Rabat, Morocco. Int J Cancer 75: 546–554. 24. Walboomers JMM, Melkert PWJ, van den Brule AJC, Snijders
5. Chichareon S, Herrero R, Munoz N, et al. (1998) Risk factors for PJF, Meijer CJLM (1992) The polymerase chain reaction for
cervical cancer in Thailand: a case–control study. J Natl Cancer human papillomavirus screening in diagnostic cytopathology of
Inst. 90: 50–57. the cervix. In Herrington CS, McGee O, eds. Diagnostic molecular
6. Eluf-Neto J, Booth M, Munoz N, Bosch FX, Meijer CJ, pathology: a practical approach. Oxford: Oxford University Press,
Walboomers JM (1994) Human papillomavirus and invasive pp. 152–172.
cervical cancer in Brazil. Br J Cancer 69: 114–119. 25. Ferlay J, Bray F, Pisani P, Parkin DM (2001) Globocan 2000:
7. Munoz N, Bosch FX, de Sanjose S, et al. (1992) The causal link Cancer Incidence, Mortality and Prevalence Worldwide. Lyon:
between human papillomavirus and invasive cervical cancer: a IARC Press.
population-based case–control study in Colombia and Spain. Int 26. Winkelstein Jr W, Shillitoe EJ, Brand R, Johnson KK (1984)
J Cancer 52: 743–749. Further comments on cancer of the uterine cervix, smoking, and
8. Ngelangel C, Munoz N, Bosch FX, et al. (1998) Causes of cervical herpesvirus infection. Am J Epidemiol 119: 1–8.
cancer in the Philippines: a case–control study. J Natl Cancer Inst 27. Winkelstein Jr W. (1990) Smoking and cervical cancer–current
90: 43–49. status: a review. Am J Epidemiol 131: 945–957.
9. Rolon PA, Smith JS, Munoz N, et al. (2000) Human papilloma- 28. Phillips AN, Smith GD (1994) Cigarette smoking as a potential
virus infection and invasive cervical cancer in Paraguay. Int cause of cervical cancer: has confounding been controlled? Int J
J Cancer 85: 486–491. Epidemiol 23: 42–49.
10. Santos C, Munoz N, Klug SJ, et al. (2001) HPV types and 29. zur Hausen H (1976) Condylomata acuminata and human genital
cofactors causing cervical cancer in Peru. Br J Cancer 85: 966–971. cancer. Cancer Res 36: 794.
11. Bosch FX, Munoz N, de Sanjose S, et al. (1993) Human 30. Herrero R, Brinton LA, Reeves WC, et al. (1989) Invasive cervical
papillomavirus and cervical intraepithelial neoplasia grade III/ cancer and smoking in Latin America. J Natl Cancer Inst 81: 205–
carcinoma in situ: a case–control study in Spain and Colombia. 211.
Cancer Epidemiol Biomarkers Prev 2: 415–422. 31. Hakama M, Luostarinen T, Hallmans G, et al. (2000) Joint effect
12. Munoz N, Bosch FX, de Sanjose S, et al. (1993) Risk factors for of HPV16 with Chlamydia trachomatis and smoking on risk of
cervical intraepithelial neoplasia grade III/carcinoma in situ in cervical cancer: antagonism or misclassification (Nordic countries).
Spain and Colombia. Cancer Epidemiol Biomarkers Prev 2: 423–431. Cancer Causes Control 11: 783–790.
13. Moreno V, Bosch FX, Munoz N, et al. (2002) Effect of oral 32. Olsen AO, Dillner J, Skrondal A, Magnus P (1998) Combined
contraceptives on risk of cervical cancer in women with human effect of smoking and human papillomavirus type 16 infection in
papillomavirus infection: the IARC multi-centric case control cervical carcinogenesis. Epidemiology 9: 346–349.
study. Lancet 359: 1085–92. 33. Ylitalo N, Sorensen P, Josefsson A, et al. (1999) Smoking and oral
14. Munoz N, Franceschi S, Bosetti C, et al. (2002) The role of parity contraceptives as risk factors for cervical carcinoma in situ. Int J
and human papillomavirus in cervical cancer: the IARC multi- Cancer 81: 357–365.
centric case–control study. Lancet 359: 1093–1101. 34. Becker TM, Wheeler CM, McGough NS, et al. (1994) Cigarette
15. Smith JS, Herrero R, Bosetti C, et al. (2002) Herpes simplex virus- smoking and other risk factors for cervical dysplasia in south-
2 as a human papillomavirus cofactor in the etiology of invasive western Hispanic and non-Hispanic white women. Cancer Epi-
cervical cancer. J Natl Cancer Inst 94: 1604–1613. demiol Biomarkers Prev 3: 113–119.
16. Castellsague X, Bosch FX, Munoz N, et al. (2002) Male circum- 35. Kjellberg L, Hallmans G, Ahren AM, et al. (2000) Smoking, diet,
cision, penile human papillomavirus infection, and cervical cancer pregnancy and oral contraceptive use as risk factors for cervical
in female partners. N Engl J Med 346: 1105–1112. intra-epithelial neoplasia in relation to human papillomavirus
17. Winkelstein Jr W. (1977) Smoking and cancer of the uterine cervix: infection. Br J Cancer 82: 1332–1338.
hypothesis. Am J Epidemiol 106: 257–259. 36. Deacon JM, Evans CD, Yule R, et al. (2000) Sexual behaviour and
18. Brinton LA (1990) Editorial commentary: smoking and cervical smoking as determinants of cervical HPV infection and of CIN3
cancer – current status. Am J Epidemiol 131: 958–960. among those infected: a case–control study nested within the
19. Layde PM (1989) Smoking and cervical cancer: cause or coinci- Manchester cohort. Br J Cancer 83: 1565–1572.
dence? JAMA 261: 1631–1633. 37. Hildesheim A, Herrero R, Castle PE, et al. (2001) HPV co-factors
20. Moreno V, Munoz N, Bosch FX, et al. (1995) Risk factors for related to the development of cervical cancer: results from
progression of cervical intraepithelial neoplasm grade III to a population-based study in Costa Rica. Br J Cancer 84: 1219–
invasive cervical cancer. Cancer Epidemiol Biomarkers Prev 4: 1226.
459–467. 38. Lacey Jr JV, Frisch M, Brinton LA, et al. (2001) Associations
21. Manos MM, Wright DK, Lewis AJ, Broker TR, Wolinsky SM between smoking and adenocarcinomas and squamous cell carci-
(1989) The use of polymerase chain reaction amplification for the nomas of the uterine cervix (United States). Cancer Causes Control
detection of genital human papillomaviruses. In: Furth M, Greaves 12: 153–161.
814 M. Plummer et al.
39. Ho GY, Kadish AS, Burk RD, et al. (1998) HPV 16 and cigarette correlation with human papillomavirus type 16, using exfoliated
smoking as risk factors for high-grade cervical intra-epithelial cervical cells. Br J Cancer 71: 246–249.
neoplasia. Int J Cancer 78: 281–285. 46. Cerqueira EM, Santoro CL, Donozo NF, et al. (1998) Genetic
40. Szarewski A, Jarvis MJ, Sasieni P, et al. (1996) Effect of damage in exfoliated cells of the uterine cervix. Association and
smoking cessation on cervical lesion size. Lancet 347: 941– interaction between cigarette smoking and progression to malig-
943. nant transformation? Acta Cytol 42: 639–649.
41. Rous P, Friedwald W (1944) The effect of chemical carcinogens on 47. Poppe WA, Ide PS, Drijkoningen MP, Lauweryns JM, Van Assche
virus-induced rabbit papillomas. J Exp Med 79: 511–538. FA (1995) Tobacco smoking impairs the local immunosurveillance
42. Yang X, Jin G, Nakao Y, Rahimtula M, Pater MM, Pater A in the uterine cervix. An immunohistochemical study. Gynecol
(1996) Malignant transformation of HPV 16-immortalized Obstet Invest 39: 34–38.
human endocervical cells by cigarette smoke condensate and 48. IARC 2003. IARC Monographs on the Evaluation of Carcinogenic
characterization of multistage carcinogenesis. Int J Cancer 65: 338– Risks to humans, Volume 83. Tobacco Smoke and Involuntary
344. Smoking. Lyon, International Agency for Research on Cancer, (in
43. IARC (1986) IARC Monographs on the Evaluation of Carcinogenic press).
Risks to Humans, Volume 38. Tobacco Smoking. Lyon: Interna- 49. Dobson AJ, Kuulasmaa K, Moltchanov V, et al. (1998) Changes
tional Agency for Research on Cancer. in cigarette smoking among adults in 35 populations in the mid-
44. Szarewski A, Cuzick J (1998) Smoking and cervical neoplasia: a 1980s. WHO MONICA Project. Tob Control 7: 14–21.
review of the evidence. J Epidemiol Biostatist 3: 229–256. 50. Ernster V, Kaufman N, Nichter M, Samet J, Yoon SY (2000)
45. Simons AM, Mugica vH, Rodriguez JA, et al. (1995) Demonstra- Women and tobacco: moving from policy to action. Bull World
tion of smoking-related DNA damage in cervical epithelium and Health Organ 78: 891–901.

You might also like