Cáncer de Pancreas

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 8

PART 6 Pancreatic Disease

SECTION II Neoplastic
C. Malignant Tumors

CHAPTER 61
Pancreatic cancer: epidemiology
Theresa Pluth Yeo and Charles J. Yeo

OVERVIEW cases (Lowenfels et al, 2004). The difference in cigarette


smoking rates in men and women narrowed in the mid-1980s.
The pancreas is an organ located in the retroperitoneum, which The stabilization of incidence rates by gender may indicate that
has both exocrine and endocrine functions. The exocrine the health consequences of smoking by women over the last 3
pancreas is composed of duct cells and acinar cells that produce decades have gained parity. Currently, cigarette smoking in the
enzymes that break down carbohydrates, proteins, and lipids United States varies by race, with 21% of white men identifying
promoting digestion. Greater than 90% of malignant pancreatic as smokers, compared with 19% of white women, 22% of
neoplasms are presumed to arise from the exocrine component African-American men and 15% of African-American women,
(Cowgill et al, 2003; Li, 2003). 17% of Hispanic men and 8% of Hispanic women, and 17%
Pancreatic ductal adenocarcinoma (PDAC) is the most of Asian men and 5% of Asian women (ACS, 2014).
common type of pancreatic cancer (PC), representing greater In the United States, the peak incidence of PDAC occurs
than 90% of the exocrine pancreas cases (see Chapter 59). The in the seventh and eighth decades of life (Howlader et al,
other 10% of exocrine tumors are mucinous tumors, mixed 2014). The U.S. population risk for pancreatic cancer is 14.0
adenosquamous tumors, or acinar cell tumors (Cowgill et al, per 100,000 for men and 10.7 per 100,000 for women. The
2003; Li, 2003). Endocrine neoplasms (islet cell tumors and main risk factor for PDAC is advancing age. Between 2007 and
pancreatic neuroendocrine tumors) contribute approximately 2011, the median age at diagnosis for cancer of the pancreas
5% of the total pancreatic cancer cases (Cowgill et al, 2003; Li, was 71 years, and the median age at death was 73 years accord-
2003). ing to SEER data (Howlader et al, 2014). More than 60% of
PDAC is responsible for 2.8% of all new cancers and 6% of new PDAC cases occur between the ages 65 and 84 years. The
all cancer deaths in the United States (Siegel et al, 2014). PC incidence of PDAC remains highest for black Americans, with
is the fourth leading cause of cancer death in men, after lung, rates of 17.2 per 100,000 in men and 14.2 per 100,000 in
prostate, and colorectal cancer, and as of 2014, it is also the women. The lowest risk of pancreatic cancer is among Asian/
fourth leading cause of cancer death in women, following lung, Pacific Islander men and women (10.7 and 8.9 per 100,000,
breast, and colorectal cancer (Siegel et al, 2014). More than respectively).
46,000 incident cases of PC are predicted annually in the Between 1950 and 1995, the worldwide incidence rates of
United States., with about 40,000 deaths. The diagnosis of PDAC increased ninefold in men (1.4 to 12.5 per 100,000) and
PDAC continues to confer an unfavorable prognosis, with an women (0.8 to 6.8 per 100,000), but rates have leveled off since
increase in the death rate for PDAC cases of 0.4% between 1985 (Lin et al, 1998).The annual incidence rate worldwide for
2006 and 2010 (American Cancer Society [ACS], 2014). all histologic types of pancreatic cancer is approximately 4.9
PDAC is predicted to become the second leading cause of new cases per 100,000 persons, ranking 12th among all cancers
cancer death by 2030 (ACS, 2014; Smith et al, 2009). This is globally (Globocan, 2012a). In 2012, 337,872 new cases of
mainly due to greatly improved survival for the other leading pancreatic cancer were reported worldwide, with 330,372
cancers and limited advances in the chemotherapeutic adjuvant annual deaths (Globocan, 2012b).
treatment of PDAC.
Traditionally, PDAC had been considered a predominantly RACE AND ETHNICITY
male cancer. Since 2000, the incidence rates in men and women
have been increasing at the about the same rate, 1.3% per year. African Americans
In 2009, the actual number of incident cases in women (21,420) African Americans have the highest death rates from cancer and
exceeded the number of new cases in men (21,050) (Jemal the shortest survival period of all racial groups (DeSantis et al,
et al, 2009). The 2014 Surveillance Epidemiology and End 2013). African-American males have higher incidence rates for
Results (SEER) data indicate that the number of new cases and all cancers combined and also for the most common cancers
incidence rates in men (23,530) and women (22,890) are nearly (prostate, lung, colorectal, and pancreas). Death rates, per
equal (Siegel et al, 2014). Cigarette smoking is the leading 100,000 population, from PDAC are 15.5 for African-American
environmental cause of PDAC, accounting for 25% to 35% of men and 12.6 for women, compared with 12.4 for white men

971
972 PART 6 PANCREATIC DISEASE Section II Neoplastic

and 9.3 for African American women. African Americans of uncovered. The etiology of PC is multifactorial and involves
lower socioeconomic status are more likely to be diagnosed with several common underlying pathways: insulin resistance,
advanced stage pancreatic cancer and are less likely to undergo inflammation, hemostasis, and infection (Maisonneuve et al,
surgery or to receive chemotherapy (Chang et al, 2005; Elou- 2015). Pancreatic carcinoma is also characterized by multiple
beidi et al, 2006; Silverman et al, 2002). Other known risk germline and somatic genetic mutations (Hruban et al, 1998).
factors for PDAC (obesity, diabetes, and pancreatitis) are more (This topic is covered extensively in Chapter 9B.) The majority
common in African Americans. of PCs (>80%) are due to sporadically occurring mutations. It
It has been reported that serum cotinine levels, the primary is estimated that less than 3% of pancreatic cancers are truly
metabolite of nicotine, are consistently higher in African Ameri- hereditary and due to inherited germline mutations and their
cans than in whites and Mexican Americans, even after adjust- respective syndromes (Wang et al, 2009).
ment for the number of cigarettes smoked per day, the number Personal risk factors for PC include tobacco exposure,
of smokers in the home, the number of hours of environmental including cigarette, cigar, and pipe smoking; ETS exposure,
tobacco smoke (ETS) exposure at work, the number of rooms also known as second-hand smoke or passive smoke exposure;
in the home, and the region of the country where the subject exposure to occupational and environmental carcinogens;
lived (Caraballo et al, 1998). This suggests that genetic differ- African-American race; Ashkenazi Jewish heritage; high-fat and
ences in cigarette product metabolism may influence rates of high-cholesterol diets; obesity; alcohol abuse; pancreatitis;
PDAC. diabetes; blood group subtype; and infectious agents (Table
61.1) (Everhardt et al, 1995; Gold et al, 1998; Maisonneuve
Ashkenazi Jews et al, 2014; Silverman et al, 1994; Wynder, 1975). Maison-
Pancreatic cancer occurs in excess in Jews, particularly Jews of neuve and Lowenfels recently (2015) completed an extensive
Ashkenazi heritage (Lynch et al, 1996). The age-standardized meta-analysis of 117 studies on putative causative factors for
rates for PDAC in Jews in Israel significantly exceed the rates PC and have calculated the population attributable risk for
for Israeli non-Jews (7.2 per 100,000 for all Jewish men and many of the known risk factors.
5.7 per 100,000 for Jewish women vs. 4.0 per 100,000 for
non-Jewish men and 2.9 per 100,000 for non-Jewish women).
Approximately 5% to 10% of this discrepancy is attributed to
the BRCA2 mutation, 6174delT, which is also associated with TABLE 61.1 Risk Factors Associated With Cancer of the
breast, gastric, ovarian, and bile duct cancers. The 6174delT Pancreas: Summary of Previous Reports
mutation is present in approximately 1% of Ashkenazi Jews
and in approximately 4% of all patients with PDAC (Ferrone Lifestyle and Environmental Dietary Factors
Factors High fat/cholesterol
et al, 2009; Struewing et al, 1995). The BRCA1 (185delAG, Overweight and obesity
Cigarette smoking (dose-
5382insC) founder mutation occurs less commonly than the response relationship) Nitrosamines in food
BRCA2 mutation. In a study of 187 Jewish patients with PC, a Environmental tobacco smoke Occupational Exposure to
BRCA1 founder mutation was identified in 1.3% of the patients exposure, particularly early Carcinogens
compared with the BRCA2 mutation, which was present in in life Asbestos, 2-naphthylamine,
Alcohol benzidine, gasoline
4.1% of the patients (Ferrone et al, 2009). Residential radon products, PAHs, dry-
Race/Ethnic Factors cleaning agents, DDT, radon
Asians African-American men and Selected High-Risk
Asian patients with PDAC have been reported to have less women Occupations
aggressive tumors than non-Asians (either black or white Native female Hawaiians Dry cleaning or chemical plant
Ashkenazi Jews work, sawmill work,
patients) and higher survival rates when assessed on a stage-
Inherited Predisposition electrical equipment
adjusted basis (Clegg et al, 2008). To determine whether Asians Hereditary pancreatitis manufacturing work, mining
develop a histologically different type of PDAC than Western Hereditary nonpolyposis and metal working
populations, Longnecker and colleagues (2000) conducted a colorectal cancer occupations
population-based study using three SEER geographic areas: Hereditary breast and ovarian Height
cancer RR, 1.81 (CI, 1.31 to 2.52)
Hawaii, San Francisco, and Seattle. They compared PC cases Familial atypical multiple mole when comparing tallest-
in Japanese, Chinese, Filipino, Hawaiian, black, and white melanoma syndrome and shortest-height
patients in these areas. The study revealed that Japanese patients Peutz-Jeghers syndrome categories for men and
had the highest fraction of localized tumors with the lowest Ataxia-telangiectasia women
grade, and that Chinese, Filipino, and Japanese women had Fanconi anemia Previous Surgery
Cystic fibrosis Cholecystectomy
longer survival times than did whites, although survival time Medical Conditions Gastrectomy
was significantly different for Japanese women only. The reason Chronic pancreatitis
for these differences is unknown, but race-related genetic and Cirrhosis
environmental exposure factors may be the cause of these Diabetes (or impaired fasting
survival discrepancies. blood glucose)
Helicobacter pylori infection
Periodontal disease
RISK FACTORS FOR PANCREATIC DUCTAL CI, Confidence interval; DDT, dichlorodiphenyl-trichloroethane; PAHs, polyaromatic hydrocarbons;
ADENOCARCINOMA AND PANCREATIC RR, relative risk.
CANCERS (SEE CHAPTER 9C) Not all listed factors have been definitively proven to cause pancreatic cancer.
Data from Ahlgren, 1996; Efthimiou et al, 2001; Ekbom et al, 1994; Garabrant et al, 1993; Gold
The risk factors for developing pancreatic cancer have been et al, 1998; Hruban et al, 1998; Kogevinas et al, 2000; Lowenfels et al, 1997; Lynch et al, 1996;
extensively studied, and new information continues to be Maisonneuve, 2015; Michaud et al, 2001; Silverman et al, 1998; and Yeo et al, 2009.
C. Malignant Tumors Chapter 61 Pancreatic cancer: epidemiology 973

than 5% of the familial aggregation of PC. The mean age of


Familial Pancreatic Cancer and Inherited onset of familial PC is similar to that of nonfamilial cases: 65.8
Genetic Disorders years versus 65.2 years (Hruban et al, 1998). Familial cases
Family history of PDAC is a strong risk factor in 5% to 10% have also been noted to have a somewhat increased incidence
of cases (Lowenfels et al, 2005). In the National Familial of secondary primary cancers (23.8%) when compared with
Pancreas Tumor Registry at Johns Hopkins Hospital, pro- their nonfamilial counterparts (18.9%).
spective studies of family members of pancreatic cancer kin-
dreds found a twofold increased risk of pancreatic cancer in Hereditary Pancreatitis
the first-degree relatives of persons with sporadic pancreatic Children and adolescents with hereditary pancreatitis (HP)
cancer and a ninefold increased risk in first-degree relatives may develop severe pancreatitis at a young age, often in child-
of those with familial pancreatic cancer (Klein et al, 2004; hood or adolescence, with a resultant 50-fold to 80-fold
Wang et al, 2009). Germline mutations of BRCA2 are found increased risk of PC developing over their lifetime. HP results
in 6% to 19% of familial pancreatic cancer patients (Murphy from germline or new somatic mutations in the PRSS1 cationic
et al, 2002). When a more stringent definition of familial trypsinogen gene (Lowenfels et al, 1997). Approximately 40%
pancreatic cancer is used, a 57-fold increased risk of PC was of those with HP will develop PC when the additional risk
reported in kindreds with three or more family members factor of cigarette smoking is added. The risk of cancer seems
affected with PC (Tersmette et al, 2001). This corresponds to to be limited to pancreatic cancers and not tumors in other
a notably high incidence rate of 301 cases per 100,000 per organs.
year compared with the SEER age-adjusted rate for the entire
U.S. population of 8.8 cases per 100,000 per year. A recent Hereditary Nonpolyposis Colorectal Cancer
report identified a germline truncating mutation of the Hereditary nonpolyposis colorectal cancer is an autosomal
PALB2 gene in 3% of familial pancreatic cancer patients dominant inherited disease that predisposes affected persons to
(Blackford et al, 2009; Jones et al, 2009). Despite these many colorectal cancer and PC (Hruban et al, 2002). It is usually
advances in the understanding of the mutations involved in caused by germline mutations in a number of DNA mismatch
the pathogenesis of PC, no single “pancreatic cancer gene” repair genes. Of the inherited syndromes associated with an
has been identified. increased risk of PDAC, hereditary nonpolyposis colorectal
Two recent studies have documented an increased risk of cancer is the least strongly linked to pancreatic cancer.
PC in persons with A, B, and AB blood types. (Amundadottir
et al, 2009; Petersen et al, 2010). This risk has been attributed Hereditary Breast and Ovarian Cancer
to an ABO single nucleotide polymorphism, rs505922. Overall, Patients with germline BRCA2 mutations have up to a 10-fold
56% of the population has a non-O blood group and the pro- increased risk (range, 3.5- to 10-fold) of PC, even in patients
portion of PDAC due to non-O blood group is 13% to 19% without a strong family history of breast cancer. Goggins and
(Maisonneuve et al, 2015). colleagues (1996) identified germline BRCA2 mutations in 7%
of sporadic (nonfamilial) PC patients screened, none of whom
Six Genetic Syndromes Associated With Pancreatic had a family history of breast cancer or PC. BRCA1 mutation
Ductal Adenocarcinoma carriers followed in a breast and ovarian cancer evaluation
Six genetic familial syndromes and their respective predisposing center, had a threefold increased risk of PC, a twofold increased
genes have been identified and linked to the development of risk for colon cancer, a fourfold increased risk for gastric cancer
PC; these are described briefly later (Table 61.2). Although and a 120-fold increased risk for fallopian tube cancer com-
individuals with these syndromes have an increased risk of pared with SEER population-based estimated risk (Brose et al,
developing PC, collectively these syndromes account for less 2002).

Familial Atypical Multiple Mole and Melanoma Syndrome


Familial atypical multiple mole and melanoma (FAMMM)
TABLE 61.2 High-Risk Genetic Disorders Associated With syndrome is rare condition associated with p16 germline muta-
tions, a tumor suppressor gene that may be mutated or may
Familial Pancreatic Cancer
have its expression altered by posttranscriptional methylation.
Gene/Chromosomal Estimated Increased The syndrome predisposes affected individuals to melanomas,
Genetic Syndrome Mutation Region Risk of PDA multiple nevi, atypical nevi, and PC (Lynch et al, 1990). Those
Hereditary PRSS1 (7q35) 50-80 times with FAMMM have a 20- to 34-fold increased risk of PC over
pancreatitis their lifetime.
Hereditary hMSH2, hMSH1, Undefined
nonpolyposis hPMS2, hMSH3, Peutz-Jeghers Syndrome
colorectal cancer hPMS1, hMSH6/ Peutz-Jeghers syndrome is a rare autosomal dominant disease
(Lynch II variant) GTBP associated with alterations in the STK11 gene, in which
Hereditary breast and BRCA2 (13q12- 3.5-10 times affected individuals develop hamartomatous polyps of the
ovarian cancer q13)
gastrointestinal tract and lip freckles referred to as mucocutane-
FAMMM syndrome p16 (9p21) 20-34 times ous melanocytic macules (Hruban et al, 2002). Individuals with
Peutz-Jeghers STK11/LKB1 75-132 times this syndrome have a roughly 100-fold increased risk of
syndrome (19p13)
developing PC (Giardiello et al, 2000), and they also appear
Ataxia-telangiectasia ATM (11q22-23) Undefined
to have a tendency to form intraductal papillary mucinous
FAMMM, Familial atypical multiple mole melanoma; PDA, pancreatic ductal carcinoma. neoplasms.
974 PART 6 PANCREATIC DISEASE Section II Neoplastic

and 14% of high school students in the United States report


Ataxia-Telangiectasia current smoking Centers for Disease Control and Prevention.
Ataxia-telangiectasia is an autosomal recessive inherited disor- A 2003 study of adults aged 30 to 39 years that included
der associated with ATM gene mutations, in which affected parental occupation, adult educational attainment, and
persons present with cerebellar ataxia, conjunctival telangiecta- household income as indicators of socioeconomic status
sias, a hypofunctioning thymus gland, and oculomotor abnor- (SES) found that those of lower SES were more likely to
malities. An association between ataxia-telangectasia and the start smoking and to become regular smokers and were less
subsequent development of PC has been reported, but it is less likely to quit smoking than their higher SES counterparts
well established than with the other five familial syndromes (Gilman et al, 2003).
(Lynch et al, 1996). The carcinogenic components of cigarette smoke are
usually cleared from the bloodstream and excreted into bile;
Tobacco Exposure they reach the pancreas from the bile duct through biliary-
Cigarette smoking has been definitively identified as a causative pancreatic reflux, or they may be carried into the pancreatic
agent in 25% to 35% of the cases of PDAC and is the most parenchyma directly by the bloodstream. Cigarette smoke
consistently reported risk factor (Iodice et al, 2008; Jemal et al, contains more than 60 known carcinogens, including polycy-
2009, Lowenfels et al, 2004). Smoking is associated with clic aromatic hydrocarbons (PAHs), nitrosamines, benzo[a]
increased risk of PC in at least 29 epidemiologic studies (Silver- pyrene, β-napthylamine, methylfluoranthenes, and arylamines
man et al, 1994), and smokers have a 70% increased risk of PC (Hecht, 2003). These carcinogens may bind to DNA and form
compared with nonsmokers. Smoking habits in the 15 years adducts, which increase the risk of somatic mutations and
preceding the diagnosis of PC appear to be more relevant to cancer if unrepaired. Nitrosamines are known to be organ
increased risk, whereas former smokers who have quit for more specific, causing pancreatic carcinomas in hamsters that are
than 13 years decrease their PC risk to that of lifetime non- histologically similar to the type found in humans (Wang et al,
smokers (Howe et al, 1991). A meta-analysis of 83 epidemio- 2009). Little research has been conducted on the other car-
logic studies found that the overall relative risk of PC in current cinogens in cigarette smoke and their relationship to PC.
and former smokers was 1.74, a 74% increased risk of PC in Another possible mechanism by which cigarette smoke leads
these groups (Iodice et al, 2008). A retrospective analysis to PC is unrelated to the carcinogens in the smoke. Rather,
comparing smoking in cases of familial PC and sporadic PC higher cholesterol (lipid) levels are measured in smokers than
found that 57% and 60%, respectively, of patients reported in nonsmokers. The pathophysiologic mechanism of hypercho-
prolonged cigarette smoking, with an overall mean of 35 packs/ lesterolemia may be partially due to nicotinic stimulation of
year history (Yeo et al, 2009). circulating catecholamines, which increase serum cholesterol
Prospective and retrospective studies have found that the levels. Elevated lipid levels may also be causative in the induc-
risk of PC increases consistently with cigarette smoking but tion of PC (Wang et al, 2009).
inconsistently with cigar or pipe smoking (Wynder, 1975).
Cigar consumption has increased 233% between 2000 and Environmental Tobacco Smoke and Second-Hand Smoke
2011; regular cigar smokers have 4 to 10 times the risk of dying ETS is related to the development of PC, with a likely dose-
from laryngeal, oral, or esophageal cancers (and likely PC) than response relationship (Ahlgren, 1996; Villeneuve et al, 2004).
do nonsmokers (Centers for Disease Control and Prevention, A mildly elevated odds ratio (OR, 1.2.1; confidence interval
2013). [CI], 0.60 to 2.44) has been reported, suggesting a weak asso-
A dose-response relationship between PDAC and the ciation between PDAC and ETS exposure in nonsmokers who
number of cigarettes consumed has been documented in reported ETS exposure both as an adult and in childhood. The
several investigations (Ahlgren, 1996; Howe et al, 1991). For effect was more pronounced in smokers who reported ETS
current smokers who also have a family history of pancreatic exposure. Findings from a 2009 retrospective case—only analy-
cancer, the relative risk of PC has been reported to be as sis of familial and sporadic PDAC—indicate that nonsmokers
high as 8.23 (Schenk, 2001). Lowenfels and colleagues (1997) with PC who were exposed to ETS early in life (<21 years of
found that individuals with familial PC tend to smoke more age) were diagnosed with PC at a significantly younger mean
than those with sporadic PC. These observations raise the age (64.0 years) when compared with nonsmoker, non–ETS-
possibility that smoking is interactive, perhaps multiplicatively, exposed cases (66.5 years) (Yeo et al, 2009). Moreover, both
with genetic mutations known to be present in persons with cigarette smoking and ETS exposure in nonsmokers younger
familial PC. Postmortem examinations of the pancreatic ducts than 21 years are associated with a younger mean age of diag-
of smokers have found widespread ductal hyperplasia, that nosis in familial, sporadic, and Ashkenazi Jewish cases of PC.
is, lesions termed pancreatic intraepithelial neoplasia, which are In 2006, the U.S. Surgeon General determined that second-
considered to be premalignant lesions. Neugut and associates hand smoke contains at least 69 human carcinogens for which
(1995) found a relationship between cigarette smoking and the there is no safe exposure level (Office on Smoking and Health
development of PC as a second malignancy in patients with a [US], 2006). There are more than 45,000 annual deaths in the
smoking-related first malignancy, such as lung, head and neck, United States from lung cancer and heart disease in persons
or bladder cancer. This relationship is likely due to overlap- who have no personal smoking history but did report exposure
ping smoking-related genetic mutations among these different to second-hand smoke (Max et al, 2012).
malignancies.
Smoking is most common in the least educated; in 2012, Smokeless Tobacco
adults with a Bachelor’s degree were less likely to be current Smokeless tobacco includes dry snuff, chewing tobacco, snus
smokers than those adults with less education. (National (moist smokeless tobacco), and dissolvable nicotine products.
Center for Health Statistics, 2012). Eighteen percent of adults Sales of these products are increasing at a faster rate than
C. Malignant Tumors Chapter 61 Pancreatic cancer: epidemiology 975

TABLE 61.3 Occupations Associated With Excess Risk of Pancreatic Cancer


Reference Occupation Risk Estimate 95% CI

Falk et al, 1994 Chemical processor 1.22 OR NR


Chemical fertilizer 1.20 OR NR
Li et al, 1969 Chemist 10.2 MH NR
Lin et al, 1981; Falk et al, 1990 Auto mechanic (gasoline exposure) 5.1 RR if >10 years exposure NR
Service station worker 1.6 SMR NR
Machine repairer 2.45 OR NR
Lin & Kessler, 1981 Dry cleaner 2.1 RR NR
Edling et al, 1985 Leather tanner 3.1 MH (1.1-9.2)
Pickle et al, 1980 Oil refinery worker 2.11 OR (0.86-5.20)
Rockette et al, 1983 Aluminum mill worker 125.2 SMR NR
Norell et al, 1986 Floor polisher 1.3 SMR NR
Falk et al, 1990 Electrical assembler/installer/repairer 2.20 OR NR
Tolbert et al, 1992 Metalworker 2.0 SMR (0.9-3.8)
Eisen et al, 1992 Manufacturing plant worker 3.0 SMR (1.2-6.24)
Bardin et al, 1997 Assembly worker 3.0 SMR (1.0-7.5)
Rotimi et al, 1993 Foundry and engine plant workers 3.0 SMR (121-624)
Bu-Tian et al, 2001 Plant/system operator 6.1 OR (1.1-33.9)
Bu-Tian et al, 2001 Pharmacist, dietician, therapist 7.1 OR (1.8-27.5)
CI, Confidence interval; MH, Mantel-Haenszel estimate; NR, not reported; OR, odds ratio; RR, relative risk; SMR, standardized mortality ratio.

cigarettes. The use of smokeless tobacco is being promoted as


TABLE 61.4 Occupational Exposures Linked to Pancreatic
safer than cigarette smoking and as a method to quit
Cancer (PC)
smoking. There is no evidence that smokeless tobacco is an
effective method of smoking cessation. These products do, PC Risk
however, cause oral, esophageal, and pancreatic cancer, as Chemical Exposure Route Estimates
well as bone loss around teeth and nicotine addiction Methylene chloride Spray paints 1.61 OR
(Bofetta, 2008). (chlorinated Paint strippers 1.40 OR
hydrocarbons) Aerosol propellant 1.6 RR
High-Risk Occupations Metal degreasing agent
Paint/paint thinners
The evidence linking occupational exposures to PC is inconsis-
Varnish
tent, reflecting the difficulty of quantifying workplace exposure Solvents
to carcinogens and of differentiating these exposures from other Pesticides/ Corn wet-milling 1.4 OR
risk factors. A number of epidemiologic investigations have herbicides
suggested excess risk of PC in certain occupations. Definitively DDT Farming 4.8 RR
establishing certain occupations as high risk for the develop- DDD Flight attendants 4.3 RR
ment of PC is difficult because of the problems with self-
Ethylan 5.0 RR
reported exposures, lack of objective quantitative monitoring
Asbestos Industrial 3.0 OR
data, personal comorbidities, and the presence of other con-
Fertilizer Farming 1.2 OR
founding and modifying risk factors. The available studies are
summarized in Table 61.3. Cotton dust Farming 4.37 OR
Cement Construction work 1.28 OR
Occupational Exposures (manufacturing of cement)
Studies examining specific occupational exposures that increase Lead Various 1.28 OR
a worker’s risk of developing PC have been conducted both in Metalworking fluids Machinist 2.0 OR
the United States and in Europe, particularly in the Scandina- DDD, Dichlorodiphenyl-dichlorethane; DDT, dichlorodiphenyl-trichloroethane; OR, odds ratio; RR,
vian countries. Occupational exposures linked to PC are sum- relative risk.
marized in Table 61.4. Overall, the occupational etiologic Data from references cited in Table 61.3.
fraction for PC was estimated at 12% in a meta-analysis of 20
occupational studies conducted between 1969 and 1998 (Oja-
jarvi et al, 2000). A retrospective analysis of more than 22
occupational and environmental studies reporting on people Calvert and colleagues (1989) conducted a review supported
with PC found that exposure to asbestos, pesticides and herbi- by the National Institute for Occupational Safety and Health of
cides, residential radon, coal products, welding products, and five cohort studies reporting an association between the use of
radiation were the most commonly reported exposures (Yeo metalworking fluids (MWFs) used in industrial machining and
et al, 2009). grinding operations and the development of all types of cancer,
976 PART 6 PANCREATIC DISEASE Section II Neoplastic

including PDAC (Acquavella et al, 1993; Hoppin et al, 2000; tolerance, insulin resistance, and hyperinsulinemia are involved
Porta et al, 1999; Rotimi et al, 1993; Tolbert et al, 1992). More in the etiology of PC.
than 1 million workers are exposed to MWFs according to
National Institute for Occupational Safety and Health estimates Pancreatitis
(Calvert et al, 1989). Substantial evidence was found for an Pancreatitis has been widely studied as a risk factor for PC.
increased risk of cancer at several sites, including the pancreas, The population-attributable fraction of pancreatitis to the
larynx, rectum, skin, scrotum, and bladder. MWFs contain development of PC was 1.34% (i.e., a 34% excess risk) in
a number of compounds suspected to be cancer initiators or an analysis of 10 pooled studies (Duell et al, 2012). When
promoters, including long-chain aliphatics, PAHs, nitrosa- the pancreatitis occurred more than 3 years prior to the
mines, sulfur-containing compounds, formaldehyde-releasing diagnosis of PC, the association was stronger (OR, 4.0; CI,
biocides, and heavy metals (Tolbert et al, 1992). 2.5 to 6.0). Acute pancreatitis may be the initial clinical
presentation of PDAC, preceding the malignant diagnosis by
Diabetes weeks or months (see Chapter 55). A 2014 Veterans Health
Diabetes and prediabetes are risk factors for PC and may also Administration retrospective study found that 10.7% of
be a consequence of PC (Liao, 2015). The majority of PDAC veterans diagnosed with PC had a history of acute pancre-
patients (50% to 80%) have either subclinical impaired glucose atitis within 2 years of the cancer diagnosis (Munigala et al,
tolerance or type 2 diabetes at the time of diagnosis (Yalniz 2014). The risk was greatest in the first year following the
et al, 2005). PC causes alteration of islet cell function— episode of acute pancreatitis and further elevated in persons
specifically, the loss of β-cell mass from tumor growth—or a 70 years of age.
result of disruptions in acinar–islet cell interactions. The chronic Chronic pancreatitis has been linked to the development of
hyperinsulinemia and hyperglycemia associated with type 2 PC (see Chapter 57). It is not clear whether chronic pancreati-
diabetes has been proposed as the underlying mechanism of tis is a risk factor or if it represents an indolent presentation of
PC. Experimental evidence suggests that insulin promotes PC. The standardized incidence ratio of PDAC in individuals
proliferation and reduces apoptosis in pancreatic cancer cells, with a diagnosis of chronic pancreatitis was 16.5 compared
both directly and indirectly through increased bioavailability of with the 1.76 expected number of cases (Lowenfels et al,
insulin-like growth factor (Butler et al, 2010; Han et al, 2011; 1993). Because PC may mimic chronic pancreatitis, misdiag-
Li, 2012). Hyperglycmia can also enhance proliferation and the nosis is a possibility. A review of the records of patients with
invasion capability of pancreatic cancer cells. A 2015 meta- acute, chronic, or unspecified pancreatitis was conducted on all
analysis of nine prospective studies involving 2408 patients inpatient medical institutions in Sweden from 1965 to 1983. In
examined the association between elevated blood glucose and 7956 patients with pancreatitis, 46 pancreatic cancers were
PC and found that every 0.56 mmol/L increase in fasting blood diagnosed compared with 21 expected cases, a standardized
glucose was associated with a 14% increase in the rate of PC incidence ratio of 2.2 (CI, 1.6 to 2.9) (Ekbom et al, 1994).
(Liao, 2015). Although it seems reasonable to conclude that the cellular
Meta-analyses performed in the 1990s on the relationship destruction and glandular dysfunction caused by pancreatitis
between diabetes and the risk of PC found that the pooled rela- may yield an environment favoring the initiation of tumor
tive risk (RR) of PDAC in people diagnosed with diabetes 5 growth, it remains problematic that some premalignant lesions
years or more prior to the PC diagnosis was double the risk such as intraductal papillary mucinous neoplasms may be ini-
(RR, 2.0; 95% CI, 1.3 to 2.2) of those without diabetes (Ever- tially incorrectly diagnosed as chronic pancreatitis.
hardt et al, 1995). In a population-based case-control study of Hereditary pancreatitis is an autosomal inherited disease
484 PDAC cases and 2099 control subjects in three geographic that begins in childhood or adolescence. A PRSS1 (7q35)
areas of the United States, those diagnosed with diabetes at mutation has been identified, and the cumulative risk of PC is
least 10 years prior to the development of PDAC had a 50% to 80% in these individuals. Cigarette smoking in combi-
50% increased risk of PDAC compared with the control nation with hereditary pancreatitis may lower the age at
group (Silverman et al, 1998). Insulin treatment did not diagnosis.
appear to alter the risk of development of PDAC (OR, 1.6 with
insulin and 1.5 without). Two other recent investigations found Cholecystectomy
a self-reported history of new onset diabetes prior to PC diag- A history of cholelithiasis and/or cholecystectomy may precede
nosis in 26% and 31% of the study populations (Yeo et al, the diagnosis of PC; however, the evidence suggesting that
2009, 2012). cholecystectomy promotes its development is weak (Maison-
During an 18-year follow-up study of 88,802 women in neuve et al, 2015). Individuals who underwent cholecystectomy
the Nurses’ Health Study (NHS), 180 incident cases of PC in the year preceding the diagnosis of PC had an exceedingly
were reported (Michaud et al, 2001). A positive association high risk of developing PC (OR, 57.9) in one investigation
was observed between fructose intake and impaired glucose (Everhardt et al, 1995). This finding is most likely explained by
metabolism and increased PC risk. The risk was most apparent the misdiagnosis of gallbladder disease, either cholelithiasis or
in women with an elevated body mass index (BMI) above cholecystitis, in the setting of cryptic PDAC, often prompting
30 kg/m2 and low physical activity. A diet high in glycemic gallbladder removal, with an undetected tumor being left in
load (defined as the glucose response to each unit of place. However, when data subset analyses were performed in
carbohydrate-containing foods), may increase the risk of PC patients with a history of cholecystectomy 20 years or more
in women with underlying insulin resistance as a result of prior to the diagnosis of PC, a 70% excess risk of PC was still
obesity. Chronically elevated plasma glucose levels and evident. In case-control and cohort studies, gastrectomy also
increased PC risk were also observed by Gapstur and col- appears to be associated with a 50% increased risk of PC
leagues (2000), further suggesting that impaired glucose (Bosetti et al, 2013; Gong et al, 2012).
C. Malignant Tumors Chapter 61 Pancreatic cancer: epidemiology 977

(hazard ratio [HR], 1.40; CI, 0.87 to 2.25; and HR, 1.60; CI,
Hormonal Factors 0.96 to 2.64, respectively) in the Finnish Cancer Registry
In recent years, a link between female reproductive factors and (Stolzenberg-Solomon et al, 2005). Fat entering the duodenum
PC has been hypothesized. A case-control study of 52 post- stimulates cholecystokinin secretion, and it is possible that
menopausal women with PDAC and 233 matched control chronic hypercholecystokininemia may be associated with an
subjects was conducted as a component of the Canadian increased susceptibility of the pancreas to carcinogens. Other
Enhanced Cancer Surveillance Project (Kreiger et al, 2001). A possible explanations include that the increased intake of satu-
questionnaire focused on reproductive history, cigarette rated fat may lead to insulin resistance and the development of
smoking, physical activity, diet, occupation, residential history, diabetes or that foods with a high soluble fat content may be
and sociodemographic information was mailed to eligible contaminated with carcinogenic organochloride compounds
patients diagnosed with PDAC. Multiparity, defined as three or from the environment.
more children, and the use of oral contraceptives were associ- Processed meats have been linked to the development of PC.
ated with a decreased risk of PDAC (OR, 0.22 and 0.36, In a prospective study of more than 200,000 people, 482
respectively). Older patient age at birth of the first child signifi- individuals developed PC during 7 years of follow-up (Nöth-
cantly increased the risk of PDAC (OR, 4.05; CI, 1.50 to 10.92 lings et al, 2007). Those who had consumed the greatest
for ages 25 to 29 years; for women 30 or older, OR, 3.78; CI, amount of processed meats had a 67% increased risk of PC.
1.02 to 14.06). No relationships were found between age of Diets laden with pork and red meat intake were associated with
menarche or menopause and PDAC. The inverse relationship a 50% increase in PC risk, but poultry, fish, dairy products, and
between parity and PDAC suggests that PDAC may be partially egg consumption conferred no additional risk. Heterocyclic
an estrogen-dependent transformation, or that estrogen may act amines and polycyclic aromatic hydrocarbons that form during
in an inhibitory manner upon pancreatic carcinogenesis. high-temperature cooking are known to be carcinogenic and
may be responsible for the increased risk of PC observed with
Lifestyle Factors and Height and Weight processed and barbecued meats. These compounds have also
Michaud and associates (2001) first reported on the relation- been linked to the development of other cancers.
ship between BMI, height, physical activity, and smoking Several recent investigations have focused on the relation-
and the risk of PC in two large, prospective cohort studies: ship between glycemic load and glycemic index. Consumption of
the Health Professional Follow-up Study (HPFS) and the fruit juices and soda has been evaluated in six studies to date
NHS. Activity levels and body weight were ascertained pro- with conflicting results (Michaud et al, 2002; Mueller et al,
spectively. A higher risk of PDAC was found among obese 2010; Nöthlings et al, 2007; Silvera et al, 2005; Stolzenberg-
men and women: 10% higher for overweight persons and Solomon et al, 2005). Two studies found no relationship
20% higher in obese persons compared with normal-weight between PDAC and glycemic load; carbohydrate intake; or
persons. A 5-unit increase in BMI corresponded to a 10% sugar, sucrose, or fructose intake (Johnson et al, 2005; Silvera
excess risk of PC (Aune, 2012). The proportion of PC et al, 2005). Three studies reported mixed findings (Michaud
attributable to obesity ranges from 3% to 16% (Maisonneuve et al, 2002; Nöthlings et al, 2007; Stolzenberg-Solomon et al,
et al, 2015). 2005). In the Multiethnic Cohort Study, BMI modified the
A direct association between above-average height and risk effect of high sucrose intake and the occurrence of PDAC, with
of PC was also observed, such that an additional 2.54 cm of elevated BMI increasing the risk and normal BMI decreasing
height above average increased the risk of PC by 6% in the the risk (Nöthlings et al, 2007). A positive association with
HPFS and by 10% in the NHS. The association between height PDAC was also observed with a high fructose intake, but no
and general cancer risk has been identified in other studies association was observed with consumption of soft drinks (diet
(Giovannucci et al, 1995; Smith et al, 1998). Maisonneuve and or regular).
colleagues (2015) found no association between height and PC. Mueller and colleagues (2010) conducted a retrospective
Height may serve as a proxy for calorie intake or exposure to analysis of the Singapore Chinese Health Study, regarding soft
growth factors, such as insulin or insulin-like growth factor-1, drink and fruit consumption and the risk of PDAC. Among
in childhood. 63,257 participants, 142 incident PDAC cases were identified,
Michaud and colleagues (2001) also reported an inverse generating an elevated HR for those consuming two to three
relationship between moderate physical activity and PC. soft drinks per week. However, only 56.4% of the PDAC cases
Walking or hiking 1.5 hours or more per week in the overweight were histologically confirmed, 5% were reported from death
cohort was associated with a 50% reduction in PC risk, but certificate data, and 38.8% were identified by signs and symp-
physical activity had no effect on PC risk for participants who toms possibly related to PDAC but not proven. The finding of
were not overweight. Likewise, BMI had no effect on risk if the an elevated HR for PDA in those who drink two to three soft
participant was a moderate exerciser. However, individuals with drinks per week is suspect in this setting and requires further
a BMI of at least 30 kg/m2 had an elevated risk of PC (RR, evaluation.
1.72; CI, 1.19 to 2.52) compared with those with a BMI of less
than 23 kg/m2. The authors speculated that the PDAC risk Coffee and Alcohol Consumption
associated with obesity might be linked to glucose intolerance, Cohort studies from the 1970s and 1980s indicating that heavy
peripheral insulin resistance, and/or hyperinsulinism. coffee and alcohol consumption led to an excess risk of PDAC
were often confounded by excessive smoking among the heavy
Diet coffee and alcohol drinkers (Michaud et al, 2001). Three sub-
It is estimated that 30% to 50% of pancreatic cancers may be sequent investigations have attempted to clarify this issue while
attributable to dietary factors. Both butter consumption and controlling for smoking (Ghadirian et al, 1991; Lagergren et al,
saturated fat intake were positively associated with PDAC 2002; Michaud et al, 2001). Ghadirian and colleagues (1991)
978 PART 6 PANCREATIC DISEASE Section II Neoplastic

conducted a case-control study of residents of greater Montreal, drinkers to 8.7 years younger for self-reported heavy alcohol
obtaining all information through questionnaires administered consumers.
to either the patient or a proxy. The results indicated that those
who consumed alcohol were, in general, at lower risk for PDAC Infectious Agents
(OR, 0.65; CI, 0.30 to 1.44). This was true for beer, white wine, Helicobacter pylori has emerged as a moderate risk factor for
and hard liquor. The OR for red wine drinkers was elevated at PDAC in analysis of seven studies (Risch, 2012). The global
1.57, but it did not achieve statistical significance. Likewise, prevalence of H. pylori infection ranges from 25% to 50% in
coffee drinkers had a lower overall risk of developing PDAC Westernized countries. It is thus estimated that 45 to 25% of
(OR, 0.55; CI, 0.19 to 1.62), regardless of whether they drank PC cases in these countries may be due to H. pylori (Maison-
caffeinated or decaffeinated coffee. The increased risk when neuve et al, 2015). Chronic infection with the hepatitis B virus
coffee was taken on an empty stomach or between breakfast (HBV) increases the risk of PDAC (OR, 1.42) in blood group
and lunch (OR, 1.30 and 1.15, respectively) was not statistically A individuals. HBV can infect the pancreas as well as the liver.
significant. Wang and colleagues (2012) reported a 40% to 60% excess risk
Lagergren and colleagues (2002) hypothesized that because of PC in HBV-positive persons.
heavy alcohol intake often causes chronic pancreatitis, which
may be a risk factor for PDAC, the ideal group to examine SUMMARY
would be those with a diagnosis of alcoholism, alcoholic chronic
pancreatitis, and alcoholic liver cirrhosis. Using the Swedish PDAC is a complex biologic process, the etiology of which is
National Board of Health and Welfare, data were collected on multifactorial, although certain risk factors are clear and ame-
178,688 patients over a 30-year period. A total of 305 incident nable to modification. Strategies to reduce PC risk include the
cases of PDA were identified in the group, representing a 40% following:
excess risk in observed compared with expected cases. Alcohol- • Avoiding the use or proximity to tobacco products,
ics with chronic pancreatitis or cirrhosis had a twofold increased specifically:
risk of PDAC. A major limitation of the study was the lack of • Do not start smoking, or if you do smoke cigarettes or
information on smoking. The authors estimated that the cigars, quit
observed excess risk of PDAC in the alcoholic group “may be • Do not use smokeless tobacco products
almost totally attributable to the confounding effect of smoking” • Avoid persons who are actively smoking
(Lagergren et al, 2002). • Ask smokers to not smoke in the home
Michaud and colleagues (2001) examined data on coffee • Diet changes
and alcohol consumption and other dietary factors obtained • If overweight or obese, reduce weight through menu-
at baseline in two large, national cohort studies. The HPFS, based and calorie restriction programs
initiated in 1986, includes 51,529 men aged 40 to 75 years • Consider surgical obesity intervention, if appropriate
who responded to a mailed questionnaire; the NHS began in • Increase daily intake of fruits and vegetables
1976 and includes 121,700 registered nurses. Follow-up • Reduce intake of simple carbohydrates and fats
information was gathered via a mailed questionnaire from • Avoid processed meats, limit red meat intake, and increase
both groups on age, marital status, weight, height, medical fish and poultry portions
history, medication use, smoking status, physical activity, and • Occupational and environmental protection
intake of coffee and alcohol. The results regarding coffee and • For those engaged in high-risk occupations or environ-
alcohol are compelling: During 1,907,222 person-years of mental exposure, regular use of the Occupational
follow-up, 288 incident cases of PDAC were diagnosed. Data Safety and Health Administration–recommended per-
were analyzed separately for each of the cohorts and then sonal protective equipment
pooled to compute overall RR estimates. The results revealed • Screening of high-risk individuals to detect precursor lesions
that neither coffee consumption nor alcohol consumption and early-stage tumors, including
conferred an excess risk of PDAC; a pooled RR of 0.62 was • Older smokers with new-onset of diabetes
reported (95% CI, 0.27 to 1.43) for more than three cups of • Unintentional weight loss
coffee per day versus no coffee, and a pooled RR of 1.00 • Suspicious pancreatic or biliary tree lesions that are
(95% CI, 0.57 to 1.76) for more than 30 g alcohol per day incidentally found and may be asymptomatic
versus no alcohol were found. A considerable amount of epidemiologic data point to PC
Most recently, Brand and colleagues (2009) examined data causes that are largely avoidable. Nearly two thirds of the major,
on 29,239 histologically confirmed cases of PC from 350 U.S. noninherited genetic risk factors are modifiable through
hospitals between the years 1993 and 2003 by cigarette smoking increased public awareness, screening of high-risk individuals
and alcohol consumption self-reported history. Current smokers to detect advanced precursor lesions, and promotion of healthier
were diagnosed with PC at significantly younger ages (6.3 to lifestyles. This knowledge offers an opportunity for prevention
8.6 years) than nonsmoking counterparts. Among nonsmokers, and perhaps bending the curve on the predicted rise of PC.
age at diagnosis of PC progressed linearly according to the
amount of alcohol consumed, from 6 years younger for minimal References are available at expertconsult.com.

You might also like