Previous reports suggest that allergic disorders may protect against various types of cancer, but the association between history of allergy and pancreatic cancer risk has not been well studied. We did a systematic review and meta-analysis of published studies to evaluate the association of any type, and specific types, of allergy and the risk of pancreatic cancer. We did a comprehensive literature search using MEDLINE, PUBMED, and the ISI Web of Science databases to identify potential relevant case-control and cohort studies. Pooled relative risks (RR) and 95% confidence intervals (95% CI) were calculated using the fixed- and random-effects model. Fourteen population-based studies (4 cohort and 10 case-control studies) with a total of 3,040 pancreatic cancer cases fulfilled our inclusion criteria. A history of allergy was associated with a reduced risk of pancreatic cancer (RR, 0.82; 95% CI, 0.68-0.99). The risk reduction was stronger for allergies related to atopy (RR, 0.71; 95% CI, 0.64-0.80), but not for asthma (RR, 1.01; 95% CI, 0.77-1.31). There was no association between allergies related to food or drugs and pancreatic cancer (RR, 1.08; 95% CI, 0.74-1.58). Overall, there was no evidence of publication bias. Allergies, in particular those related to atopy, seem to be associated with a decreased risk of pancreatic cancer. The hyperactive immune system of allergic individuals may, therefore, in some way lead to increased surveillance and protect against pancreatic cancer development.

Although the incidence of pancreatic cancer is low, because of its aggressive nature it is the fourth most common cause of death from cancer in the United States (1). Smoking is the major etiologic factor that has been linked to this lethal tumor, but only ∼25% of all pancreatic cancer are attributable to this cause (2). Furthermore, most smokers will never develop pancreatic or other types of cancer, suggesting that detoxifying mechanisms and/or the body's innate immune system protect us against cancer.

It is reasonable to assume that the immune system in allergic individuals would differ from that of nonallergic individuals, and that this difference might be responsible for quantitative differences in cancer incidence rates or in responsiveness to therapy. Indeed, several studies have suggested that the overall incidence of cancer is lower in allergic individuals than in nonallergic persons (3-12). One potential source of protection against cancer may be through increased immune surveillance in allergic individuals. The concept of immune surveillance hypothesizes that the immune system is capable of detecting and eliminating neoplastic and preneoplastic cells before they are clinically diagnosed. Many immune cell types may be involved in surveillance, but the cytokine IFN-γ system is central to the surveillance mechanism. The hyperactive immune system of allergic individuals may, therefore, in some way lead to increased surveillance.

With respect to pancreatic cancer, some studies have looked at allergy as a risk factor, but usually only as part of a comprehensive report where the primary focus has been on other risk factors, such as smoking or diet. Also, the results have been unclear because of the wide heterogeneity of terms used to define allergy. Additional uncertainty derives from the fact that, in some studies, information was obtained from proxy interviews or was based on hospital controls: These types of studies could be an important source of bias. We, therefore, did a meta-analysis and a sensitivity analysis of all published epidemiologic studies to quantitate the association between atopic allergy and pancreatic cancer.

Definition of Exposures and Outcome

The exposure variables include various types of allergy: We classified in the “atopy allergy” group patients with allergic and other types of asthma, atopic dermatitis (eczema), rhinitis (hay fever and year-around rhinitis), allergy to natural antigens, allergy to animals or plants, hives, urticaria, and reaction to insect bites and stinging insects. In subsequent analyses, we considered separately patients with (a) asthma, (b) respiratory allergy to natural antigen (including allergies to animal, plants, and dust), (c) eczema and other skin reaction (urticaria, hives, and contact dermatitis), and (d) reaction to insect bites and stinging insects. Finally, we classified in the “systemic allergy” group patients with reaction to food, medications, and chemical and commercial products that often are more irritant than allergenic.

For the outcome variable (pancreatic cancer), we relied upon the definition as published in each report.

Data Sources and Search Strategy

Published reports were obtained from the following databases using validated search strategies (13-15): Ovid MEDLINE database (1966 to July 2004); ISI Web of Science Science Citation Index Expanded (SCI Expanded); and PUBMED (http://www.ncbi.nlm.nih.gov/entrez/query.fcgi). Other sources were found in the reference lists of the retrieved articles and preceding reviews on the topic. The following search terms (both as MeSH terms and as keywords) were used to identify potentially relevant studies in the three databases mentioned above: pancreatic cancer, malignancy, atopy, atopic disease, allergy, allergic disease, asthma, eczema, hives, hay fever, and rhinitis. The search was limited to human studies but no language or time restrictions were applied.

Selection of Articles

All searches were made independently by two abstractors (S. Gandini and P. Maisonneuve); in case of disagreement or uncertainty, a third reviewer (A. Lowenfels) was consulted. Primary inclusion criteria were developed for the selection of all relevant articles (i.e., case-control, cohort, or cross-sectional studies) published as an original article. Secondary criteria were then identified to set apart studies with comparable features:

  • The studies should have sufficient information to allow adequate estimation of the relative risk (RR) and 95% confidence intervals (95% CI: i.e., they should report either adjusted odds ratios or RRs or crude data and SEs, variance, CIs, or P values of the significance of the estimates). An estimate of the RR and its variance are required to calculate a weighted pooled estimate of the RR for allergy.

  • The studies should be independent to avoid giving double weight to some estimates.

  • The populations studied should be homogeneous. In particular, when available, population-based estimates were preferred to estimates based on hospital controls and fully hospital-based studies were initially excluded. This decision has been discussed and evaluated in the sensitivity analysis.

Extraction and Classification of the Data

For each study, the following data were retrieved:

  • Study: publication year, study design, study location, mean age of study population, type of interviews.

  • Exposure: definition of the types of allergy studied.

  • Cases: number and source of cases, participation rates of cases, accrual period.

  • Controls: number and source of controls, matching design, blinding of interviewers, response rates of controls, exclusion of specific types of diseases/cancer among controls.

  • Statistics: statistical methods used, adjustment for confounding variables (demographic factors, such as age and sex, and baseline host characteristics such as smoking), type of effect estimates (odds ratio, RR, standardized incidence ratio) with corresponding measures of precision.

Statistical Methods

Because pancreatic cancer is a rare disease, we ignored the distinction between the various measures of RR (i.e., odds ratio, rate ratio, risk ratio). We transformed the various estimates of RR and their CIs into log RR and we calculated the corresponding variance using the formula proposed by Greenland (16). When estimates were not given, we calculated them from tabular data and we used Woolf's formula to evaluate the SE of the log odds ratio. When standardized incidence rates were presented, we used the number of cases to estimate the SE of the log(standardized incidence rates). Finally, “test-based” estimates were considered when only significance levels were published.

We assessed the homogeneity of the effect across studies using the large sample test based on the χ2 statistic. Because the χ2 test has limited power, we considered statistically significant heterogeneity at the P = 0.10 level of association (17). The summarized RR was estimated pooling the study-specific estimates by the classic fixed-effects and random-effects models according to the heterogeneity test. When several measures of RR were given for a single study, even if heterogeneity was not statistically significant, random-effects models were used, including the two sources of variation (within and between studies), to take into account also correlation within study. Random-effects models were fitted using SAS (Proc Mixed) with restricted maximum likelihood estimate; thus, the resulting estimate for the between-study variance is identical to the iterated DerSimonian-Laird estimator (18, 19).

We carried out subgroup analyses and meta-regression with ANOVA models to investigate between-study and between-estimates heterogeneity. We did a sensitivity analysis to evaluate the influence of various inclusions criteria and specific studies on the pooled estimates and on heterogeneity. We assessed whether publication bias might affect the validity of the estimates using two funnel-plot–based approaches: Copas and Shi sensitivity analysis (20) and the funnel plot regression of ln(RR) on the sample size, weighted by the inverse of the pooled variance (21).

Fourteen population-based studies (4 cohort and 10 case-control studies) with a total of 3,040 pancreatic cancer cases, published between 1981 and 2003, fulfilled our inclusion criteria (Table 1; refs. 2236). Six of them provided estimates partially based on proxy interviews and for two of them, separate estimates restricted to direct interviews were also given. Most studies reported estimates for several types of allergies. Only one study examined the association between atopy, determined by skin-prick testing, and cancer (ref. 33; Table 2).

Table 1.

Characteristics of studies included in the meta-analysis or sensitivity analysis

First author, year (reference)StudyCountryAccrual periodStudy subjectsHistologic confirmationType of interviewControl subjectsPair matching and adjustments
Lin, 1981 (22)* Case-control United States, 115 hospitals 1972-1975 109 cases 100% Direct 109 hospital controls Matched for age, sex, race, marital status 
Gold, 1985 (23) Case-control United States, Baltimore 1978-1980 201 cases 62% 75% proxy 201 population controls Matched on age, sex, race; adjusted for religion, alcohol, and smoking 
Mack, 1986 (24) Case-control United States, LA county 1976- 490 cases 100% 75% proxy 490 neighborhood controls Matched on age, sex, race and neighborhood 
McWhorter, 1988 (25) Cohort United States, NHANESI 1971-1975 11 incident cases Hospital records or death certificate Proxy for disabled/deceased persons 6,108 adults Adjusted for age, sex, race, and smoking 
Mills, 1988 (26) Cohort United States, California 1976-1983 40 deaths 70% Mailed questionnaire 34,000 Seventh-Day Adventists Adjusted for age and sex 
Farrow, 1990 (27) Case-control United States, Washington 1982-1986 148 married men 46% Wives 188 population controls Adjusted for age 
La Vecchia, 1990 (28) Case-control Italy, Greater Milan area 1983-1988 247 cases 100% Direct 1,089 hospital controls Adjusted for age and sex 
Jain, 1991 (29) Case-control Canada, Toronto 1983-1986 249 cases 69% 66% proxy 505 population controls Matched on age, sex, proxy status; adjusted for calories, fiber, smoking 
Bueno de Mesquita, 1992 (30) Case-control The Netherlands, central part 1984-1988 176 cases 68% 39% proxy 487 population controls Adjusted for age, sex, proxy and smoking 
Kalapothaki, 1993 (31) Case-control Greece, Athens 1991-1992 181 cases 100% Direct only 181 hospital visitors Age, sex and hospital 
Dai, 1995 (32) Case-control China, Shanghai 1992-1993 108 cases Cancer registry data Direct 275 population controls Adjusted for age, sex, income, and smoking 
Eriksson, 1995 (33) Cohort Sweden, Halmstad 1976-1989 1 incident case Cancer registry data Hospital data 6,593 patients with skin prick test Adjusted for age, sex, year 
Silverman, 1999 (34) Case-control United States, Atlanta, Detroit, New Jersey 1986-1989 484 cases 85% Direct 2,099 population controls Adjusted for age, sex, race, region, alcohol, body mass index, caloric intake, income, marital status and smoking 
Stolzenberg-Solomon, 2002 (35) Cohort Finland 1985-1988 172 cases 80% Direct 29,048 male smokers part of the α-Tocopherol, β-Carotene Cancer Prevention Study Adjusted for age, smoking, diabetes, occupation, high blood pressure 
Holly, 2003 (36) Case-control United States, San Francisco 1994-2001 532 cases 100% Direct 1,701 population controls Adjusted for age and sex and other potential confounders 
First author, year (reference)StudyCountryAccrual periodStudy subjectsHistologic confirmationType of interviewControl subjectsPair matching and adjustments
Lin, 1981 (22)* Case-control United States, 115 hospitals 1972-1975 109 cases 100% Direct 109 hospital controls Matched for age, sex, race, marital status 
Gold, 1985 (23) Case-control United States, Baltimore 1978-1980 201 cases 62% 75% proxy 201 population controls Matched on age, sex, race; adjusted for religion, alcohol, and smoking 
Mack, 1986 (24) Case-control United States, LA county 1976- 490 cases 100% 75% proxy 490 neighborhood controls Matched on age, sex, race and neighborhood 
McWhorter, 1988 (25) Cohort United States, NHANESI 1971-1975 11 incident cases Hospital records or death certificate Proxy for disabled/deceased persons 6,108 adults Adjusted for age, sex, race, and smoking 
Mills, 1988 (26) Cohort United States, California 1976-1983 40 deaths 70% Mailed questionnaire 34,000 Seventh-Day Adventists Adjusted for age and sex 
Farrow, 1990 (27) Case-control United States, Washington 1982-1986 148 married men 46% Wives 188 population controls Adjusted for age 
La Vecchia, 1990 (28) Case-control Italy, Greater Milan area 1983-1988 247 cases 100% Direct 1,089 hospital controls Adjusted for age and sex 
Jain, 1991 (29) Case-control Canada, Toronto 1983-1986 249 cases 69% 66% proxy 505 population controls Matched on age, sex, proxy status; adjusted for calories, fiber, smoking 
Bueno de Mesquita, 1992 (30) Case-control The Netherlands, central part 1984-1988 176 cases 68% 39% proxy 487 population controls Adjusted for age, sex, proxy and smoking 
Kalapothaki, 1993 (31) Case-control Greece, Athens 1991-1992 181 cases 100% Direct only 181 hospital visitors Age, sex and hospital 
Dai, 1995 (32) Case-control China, Shanghai 1992-1993 108 cases Cancer registry data Direct 275 population controls Adjusted for age, sex, income, and smoking 
Eriksson, 1995 (33) Cohort Sweden, Halmstad 1976-1989 1 incident case Cancer registry data Hospital data 6,593 patients with skin prick test Adjusted for age, sex, year 
Silverman, 1999 (34) Case-control United States, Atlanta, Detroit, New Jersey 1986-1989 484 cases 85% Direct 2,099 population controls Adjusted for age, sex, race, region, alcohol, body mass index, caloric intake, income, marital status and smoking 
Stolzenberg-Solomon, 2002 (35) Cohort Finland 1985-1988 172 cases 80% Direct 29,048 male smokers part of the α-Tocopherol, β-Carotene Cancer Prevention Study Adjusted for age, smoking, diabetes, occupation, high blood pressure 
Holly, 2003 (36) Case-control United States, San Francisco 1994-2001 532 cases 100% Direct 1,701 population controls Adjusted for age and sex and other potential confounders 
*

Excluded from the main pooled estimates.

Table 2.

Summary of published study results

First author, yearSource or type of allergyDirect + proxy interviews, RR (95% CI)Direct interview only, RR (95% CI)Classification group
Lin, 1981* Allergy; eczema; dermatitis — 2.56 (1.34-4.89) Any allergy 
Gold, 1985 Allergic disorders 0.97 (0.50-1.90) — Any allergy 
Mack, 1986 Any allergic disease 0.6 (0.4-0.8) 0.2 (0.1-0.5) Any allergy 
 Asthma 0.7 (0.4-1.1) 0.2 (0.1-0.8) Asthma 
 Eczema; hives 0.2 (0.1-0.5) 0.2 (0.03-0.7) Dermal 
 Hay fever; plants; animals 0.5 (0.3-0.9) 0.2 (0.05-0.7) Respiratory 
 Drugs; cosmetics and household products 0.9 (0.5-1.6) 0.4 (0.1-1.6) Systemic 
McWhorther, 1988 Asthma, hay fever, hives, food, or other allergies 1.69 (0.49-5.83) — Any allergy 
Mills, 1988 Asthma — 0.87 (0.21-3.63) Asthma 
 Reaction to poison ivy, oak or other plants — 0.99 (0.44-2.27) Dermal 
 Reaction to bee sting — 0.43 (0.06-3.16) Insect bites 
 Hay fever — 0.66 (0.23-1.87) Respiratory 
 Drugs — 0.67 (0.07-5.98) Systemic 
 Chemicals — 1.53 (0.37-6.30) Systemic 
Farrow, 1990 Asthma 1.1 (0.4-3.2) — Asthma 
 Plants 0.7 (0.3-1.8) — Respiratory 
 Animals 1.2 (0.4-3.4) — Respiratory 
 Drugs 1.7 (1.0-3.0) — Systemic 
 Foods 2.1 (0.8-5.5) — Systemic 
La Vecchia, 1990 Drugs — 0.94 (0.56-1.57) Systemic 
Jain, 1991 Asthma 0.52 (0.16-1.70) — Asthma 
 Eczema 0.68 (0.31-1.51) — Dermal 
 Hay fever 0.47 (0.18-1.27) — Respiratory 
 Other allergies 1.26 (0.69-2.28) — — 
Bueno de Mesquita, 1992 Any allergy 0.57 (0.36-0.90) 0.43 (0.23-0.81) Any allergy 
 Eczema 0.75 (0.42-1.32) 0.71 (0.34-1.48) Dermal 
 Asthma, hay fever, others 0.41 (0.21-0.82) 0.22 (0.08-0.63) Respiratory 
Kalapothaki, 1993 Allergic asthma — 0.50 (0.04-5.57), Asthma 
Dai, 1995 Any conditions — 0.6 (0.4-1.1) Any allergy 
 Asthma — 1.0 (0.3-3.2) Asthma 
 Contact dermatitis — 0.5 (0.1-1.7) Dermal 
 Urticaria — 0.5 (0.2-1.3) Dermal 
 Mosquito bites — 1.0 (0.3-3.1) Insect bites§ 
 Allergic rhinitis — 0.3 (0.1-1.4) Respiratory 
 Drugs — 1.1 (0.5-2.4) Systemic 
 Food — 0.3 (0.0-2.6) Systemic 
Eriksson, 1995 Positive skin prick test to inhalant allergens — 1.22 (0.03-6.80) Respiratory 
Silverman, 1999 Any allergic condition — 0.7 (0.5-0.9) Any allergy 
 Asthma — 1.0 (0.6-1.5) Asthma 
 Eczema — 1.1 (0.7-1.9) Dermal 
 Insect bite/sting — 0.8 (0.6-1.2) Insect bites 
 Hay fever — 0.6 (0.5-0.9) Respiratory 
 Animals — 0.5 (0.2-1.1) Respiratory 
 Dust or mold — 0.6 (0.3-1.1) Respiratory 
 Drugs — 1.4 (1.0-1.9) Systemic 
 Household products — 1.5 (0.8-2.9) Systemic 
Stolzenberg-Solomon, 2002 Bronchial asthma — 2.16 (1.17-3.98) Asthma 
 Allergic skin lesions — 0.59 (0.29-1.20) Dermal 
Holly, 2003 Eczema — 0.66 (0.46-0.93) Dermal 
 Other allergies — 0.77 (0.63-0.95) — 
 Insect bites or stings — 0.65 (0.41-1.00) Insect bites 
 House dust — 0.72 (0.54-0.94) Respiratory 
 Plants — 0.77 (0.62-0.96) Respiratory 
 Mold — 0.49 (0.32-0.75) Respiratory 
 Any animals — 0.66 (0.47-0.93) Respiratory 
 Food — 0.74 (0.51-1.10) Systemic 
First author, yearSource or type of allergyDirect + proxy interviews, RR (95% CI)Direct interview only, RR (95% CI)Classification group
Lin, 1981* Allergy; eczema; dermatitis — 2.56 (1.34-4.89) Any allergy 
Gold, 1985 Allergic disorders 0.97 (0.50-1.90) — Any allergy 
Mack, 1986 Any allergic disease 0.6 (0.4-0.8) 0.2 (0.1-0.5) Any allergy 
 Asthma 0.7 (0.4-1.1) 0.2 (0.1-0.8) Asthma 
 Eczema; hives 0.2 (0.1-0.5) 0.2 (0.03-0.7) Dermal 
 Hay fever; plants; animals 0.5 (0.3-0.9) 0.2 (0.05-0.7) Respiratory 
 Drugs; cosmetics and household products 0.9 (0.5-1.6) 0.4 (0.1-1.6) Systemic 
McWhorther, 1988 Asthma, hay fever, hives, food, or other allergies 1.69 (0.49-5.83) — Any allergy 
Mills, 1988 Asthma — 0.87 (0.21-3.63) Asthma 
 Reaction to poison ivy, oak or other plants — 0.99 (0.44-2.27) Dermal 
 Reaction to bee sting — 0.43 (0.06-3.16) Insect bites 
 Hay fever — 0.66 (0.23-1.87) Respiratory 
 Drugs — 0.67 (0.07-5.98) Systemic 
 Chemicals — 1.53 (0.37-6.30) Systemic 
Farrow, 1990 Asthma 1.1 (0.4-3.2) — Asthma 
 Plants 0.7 (0.3-1.8) — Respiratory 
 Animals 1.2 (0.4-3.4) — Respiratory 
 Drugs 1.7 (1.0-3.0) — Systemic 
 Foods 2.1 (0.8-5.5) — Systemic 
La Vecchia, 1990 Drugs — 0.94 (0.56-1.57) Systemic 
Jain, 1991 Asthma 0.52 (0.16-1.70) — Asthma 
 Eczema 0.68 (0.31-1.51) — Dermal 
 Hay fever 0.47 (0.18-1.27) — Respiratory 
 Other allergies 1.26 (0.69-2.28) — — 
Bueno de Mesquita, 1992 Any allergy 0.57 (0.36-0.90) 0.43 (0.23-0.81) Any allergy 
 Eczema 0.75 (0.42-1.32) 0.71 (0.34-1.48) Dermal 
 Asthma, hay fever, others 0.41 (0.21-0.82) 0.22 (0.08-0.63) Respiratory 
Kalapothaki, 1993 Allergic asthma — 0.50 (0.04-5.57), Asthma 
Dai, 1995 Any conditions — 0.6 (0.4-1.1) Any allergy 
 Asthma — 1.0 (0.3-3.2) Asthma 
 Contact dermatitis — 0.5 (0.1-1.7) Dermal 
 Urticaria — 0.5 (0.2-1.3) Dermal 
 Mosquito bites — 1.0 (0.3-3.1) Insect bites§ 
 Allergic rhinitis — 0.3 (0.1-1.4) Respiratory 
 Drugs — 1.1 (0.5-2.4) Systemic 
 Food — 0.3 (0.0-2.6) Systemic 
Eriksson, 1995 Positive skin prick test to inhalant allergens — 1.22 (0.03-6.80) Respiratory 
Silverman, 1999 Any allergic condition — 0.7 (0.5-0.9) Any allergy 
 Asthma — 1.0 (0.6-1.5) Asthma 
 Eczema — 1.1 (0.7-1.9) Dermal 
 Insect bite/sting — 0.8 (0.6-1.2) Insect bites 
 Hay fever — 0.6 (0.5-0.9) Respiratory 
 Animals — 0.5 (0.2-1.1) Respiratory 
 Dust or mold — 0.6 (0.3-1.1) Respiratory 
 Drugs — 1.4 (1.0-1.9) Systemic 
 Household products — 1.5 (0.8-2.9) Systemic 
Stolzenberg-Solomon, 2002 Bronchial asthma — 2.16 (1.17-3.98) Asthma 
 Allergic skin lesions — 0.59 (0.29-1.20) Dermal 
Holly, 2003 Eczema — 0.66 (0.46-0.93) Dermal 
 Other allergies — 0.77 (0.63-0.95) — 
 Insect bites or stings — 0.65 (0.41-1.00) Insect bites 
 House dust — 0.72 (0.54-0.94) Respiratory 
 Plants — 0.77 (0.62-0.96) Respiratory 
 Mold — 0.49 (0.32-0.75) Respiratory 
 Any animals — 0.66 (0.47-0.93) Respiratory 
 Food — 0.74 (0.51-1.10) Systemic 
*

Excluded from main meta-analysis.

RR, 0.25; 95% CI, 0.03-2.24 for hospital controls; RR, 0.33; 95% CI, 0.01-12.4 for all.

Estimates calculated on crude data.

§

Strong reaction to mosquito bites.

Standardized incidence rates for severe and intermediate atopy were published separately.

The pooled RR indicated a significant protective effect for “any allergy” against pancreatic cancer (RR, 0.82; 95% CI, 0.68-0.99; Fig. 1). For allergies related to atopy, the inverse association was even stronger (RR, 0.71; 95% CI, 0.64-0.80). The protective effect was present for respiratory allergy excluding asthma (RR, 0.63; 95% CI, 0.52-0.76; Fig. 2) and for dermal allergy (RR, 0.66; 95% CI, 0.49-0.89; Fig. 3) but not for asthma, which is not always related to atopy (RR, 1.01; 95% CI, 0.77-1.31; Fig. 4). Only seven studies have reported on allergies related to food or drugs, showing no association with pancreatic cancer risk (RR, 1.08; 95% CI, 0.74-1.58; Table 3). When restricting the meta-analysis to the eight studies, which provided risk estimates adjusted for smoking, the protective effect for any allergy became stronger (RR, 0.75; 95% CI, 0.65-0.87).

Figure 1.

Meta-analysis: Forest plot and pooled RR of the association between any allergy and pancreatic cancer using the random-effects model.

Figure 1.

Meta-analysis: Forest plot and pooled RR of the association between any allergy and pancreatic cancer using the random-effects model.

Close modal
Figure 2.

Meta-analysis: Forest plot and pooled RR of the association between respiratory allergy and pancreatic cancer using the random-effects model.

Figure 2.

Meta-analysis: Forest plot and pooled RR of the association between respiratory allergy and pancreatic cancer using the random-effects model.

Close modal
Figure 3.

Meta-analysis: Forest plot and pooled RR of the association between dermal allergy and pancreatic cancer using the random-effects model.

Figure 3.

Meta-analysis: Forest plot and pooled RR of the association between dermal allergy and pancreatic cancer using the random-effects model.

Close modal
Figure 4.

Meta-analysis: Forest plot and pooled RR of the association between asthma and pancreatic cancer using the random-effects model.

Figure 4.

Meta-analysis: Forest plot and pooled RR of the association between asthma and pancreatic cancer using the random-effects model.

Close modal
Table 3.

Overall estimates of RR of pancreatic cancer by subgroups

No. studies* (n)Direct + proxy interviews RR (95% CI)χ2PNo. studies* (n)Direct interviews only, RR (95% CI)χ2P
Any allergy 14 0.82 (0.68-0.99) 0.07 10 0.70 (0.51-0.97) 0.02 
    Atopic allergy 13 0.71 (0.64-0.80) 0.12 0.64 (0.43-0.95) 0.04 
        Respiratory 0.63 (0.52-0.76) 0.68 0.54 (0.32-0.82) 0.30 
        Asthma 1.01 (0.77-1.31) 0.22 0.85 (0.42-1.71) 0.01 
        Any dermal 0.66 (0.49-0.89) 0.21 0.76 (0.61-0.95) 0.68 
            Eczema, hives… 0.66 (0.42-1.03) 0.08 0.72 (0.52-0.98) 0.40 
            Insect bites 0.74 (0.57-0.97) 0.79 0.74 (0.57-0.97) 0.79 
    Systemic (drugs, food, …) 1.08 (0.74-1.58) 0.08 0.95 (0.60-1.50) 0.09 
No. studies* (n)Direct + proxy interviews RR (95% CI)χ2PNo. studies* (n)Direct interviews only, RR (95% CI)χ2P
Any allergy 14 0.82 (0.68-0.99) 0.07 10 0.70 (0.51-0.97) 0.02 
    Atopic allergy 13 0.71 (0.64-0.80) 0.12 0.64 (0.43-0.95) 0.04 
        Respiratory 0.63 (0.52-0.76) 0.68 0.54 (0.32-0.82) 0.30 
        Asthma 1.01 (0.77-1.31) 0.22 0.85 (0.42-1.71) 0.01 
        Any dermal 0.66 (0.49-0.89) 0.21 0.76 (0.61-0.95) 0.68 
            Eczema, hives… 0.66 (0.42-1.03) 0.08 0.72 (0.52-0.98) 0.40 
            Insect bites 0.74 (0.57-0.97) 0.79 0.74 (0.57-0.97) 0.79 
    Systemic (drugs, food, …) 1.08 (0.74-1.58) 0.08 0.95 (0.60-1.50) 0.09 
*

Published studies for which we have at least one estimate.

P value for heterogeneity χ2 test.

Face-to-Face versus Proxy Interviews

Because of the rapidly fatal course and extreme morbidity of pancreatic cancer, often case-control studies relied on information collected from relatives or friends (proxy respondents). It is obvious that for some lifestyle or personal history characteristics, such as allergy, data collected by proxy are likely to be less accurate and less reliable than data collected from face-to face interviews. When separate estimates for proxy and for direct interviews were available from a single study, it seemed that the inverse association between history of allergies and pancreatic cancer was stronger in direct interviews (24, 30). In fact, most pooled estimates (except for dermal allergy) decreased noticeably when restricted to direct interviews (Table 3). For any allergy, it was possible to obtain a pooled estimate based on 10 studies (RR, 0.70; 95% CI, 0.51-0.97).

Sensitivity Analysis

We did a sensitivity analysis to assess the influence of various studies or various study characteristics on the pooled estimates: Initially, we excluded the study by Lin et al. (22) from the analysis for several reasons: The study was carried out in 115 hospitals, between 1972 and 1975, but no description of the control group was given; it was not stated whether the hospital controls may have respiratory problems or diseases related to allergy and, therefore, may be subject to introduce a bias in the study results; the study was carried out before computed tomography scan and, therefore, the diagnosis of pancreatic cancer might not have been always accurate; only the number and the frequency of allergic cases and controls were given, which did not allow us to calculate adjusted estimates. After inclusion of this study, the pooled RR for any allergy lost statistical significance (RR, 0.88; 95% CI, 0.70-1.11) and heterogeneity became substantial (P = 0.005). Similar results were found for atopic allergy, with the pooled RR showing just a marginal protective effect (RR, 0.82; 95% CI, 0.62-1.07) and again with appearance of significant heterogeneity (P = 0.008), providing strong support for exclusion of the study from the main meta-analysis.

The heterogeneity observed for any allergy was driven by a single study (35) that has peculiar characteristics: This study was part of the α-Tocopherol, β-Carotene Cancer Prevention Study and was restricted to male smokers with no medical problems who might have limited their long-term participation to the trial. In this study, “bronchial asthma” (with no mention of allergy), was associated with a significant 2-fold risk for developing pancreatic cancer but it might well have been a marker of cigarette dose, as discussed by the authors. After exclusion of this study from the meta-analysis, the pooled RR for any allergy improved in significance (RR, 0.79; 95% CI, 0.65-0.95) and heterogeneity disappeared (P = 0.32). Similar results were found for atopic allergy, with the pooled RR showing a strong protective effect (RR, 0.61; 95% CI, 0.43-0.86) with no sign of heterogeneity (P = 0.29).

The cohort study by Erikkson et al. (33) concerns a very young population (median age is 31 and 90th percentile is 55 years) and is the only one using skin prick test. The authors showed that such test was negative for many subjects who declared suffering from asthma, rhinitis, or urticaria; therefore, assessment of allergy in this cohort differs from the other studies. Still, in view of the very wide CIs of the RR estimate and the very low weight of this study based on one single case of pancreatic cancer, its inclusion did not influence the pooled RR. Similarly, the study by Kalapothaki et al. (31) based on very few cases did not influence the overall estimates.

In contrast, the study by Holly et al. (36), the most recent, is also the largest one. Its estimates, which have a considerable weight on the pooled RR, derive from very detailed measures. After exclusion of this study from the meta-analysis, the pooled RR for any allergy was of borderline statistical significance (RR, 0.83; 95% CI, 0.67-1.04), whereas the estimate for atopic allergy remained similar (RR, 0.73; 95% CI, 0.59-0.91).

We did further analysis to evaluate if the inclusion of multiple estimates from a single study may have influenced the pooled RR, giving too much weight to some studies. This was not a problem for asthma, eczema, or for reactions to mosquito bites because no more than one estimate per study was available for these categories. Instead, in case of multiple estimates for a single allergy category, such as “respiratory allergy to natural antigens,” we arbitrarily choose the one that we retained to be most relevant: in that case, we preferred “allergy to plant” and “hay fever” to “allergy to animals,” “allergy to house dust,” or “allergy to mold,” which we retained less specific or less common forms of allergy. After exclusion of the multiple estimates, heterogeneity was not significant (P = 0.74) and the fixed-effects model applied did not show a considerable change (RR, 0.74; 95% CI, 0.65-0.83). This confirmed that the random-effects model applied to the main analyses, which takes into account correlation within each study, was conservative because it produced larger CIs.

Finally, the Funnel-plot–based approaches did not suggest any indication for publication bias.

Atopy and Cancer Development

Results from this meta-analysis support evidence of a reduced risk of pancreatic cancer among persons with a history of allergic conditions. The inverse association is moderate and the overall estimate of borderline significance. However, such association has been noted for several other forms of cancer: In 1960, Fisherman (37) suggested that natural defenses against cancer may explain (a) the differences between a cancer-prone and a cancer-resistant person; (b) the differences in tumor growth rate, invasiveness, and curability; (c) the occasional rare spontaneous disappearance of cancer. In his study, the prevalence of atopy was significantly lower (3.2%) among 1,185 patients with malignancy than among a control group of 294 noncancerous patients (12.9%), findings later confirmed by Vena et al. (3). Allergic conditions have been associated with reduced risk of cancer of the oral cavity, pharynx, larynx, colon and rectum (7), esophagus (7, 32), stomach (4), breast (4, 12), malignant melanoma, body of the uterus (4), glioma (5, 6), multiple myeloma (4), acute myelocytic leukemia (8), childhood acute lymphoblastic leukemia (9, 10), and non–Hodgkin's lymphoma (11). Still, in several studies, the authors were not able to identify such risk reduction (3842). Mills et al. (43), who found elevated risk of prostate and breast cancer but decreased risk of ovarian cancer in person who reported any type of allergic history, concluded that the association between allergy and cancer is complex and depends on the specific allergy and the specific organ site under consideration.

Atopy, Natural T Cells, and Prognosis of Cancer Patients

Several studies have also shown the importance of tumor immunity on cancer prognosis. Recently, Pompei et al. (44) not only found that the prevalence of allergy was lower in a series of 1,055 consecutive cancer patients than in a control group (8% versus 16-37%), but that allergic patients had a 20% higher probability of being cured and a 50% lower risk of tumor progression compared with nonallergic patients, suggesting that allergy-related overactive immune system is associated with cancer prognosis. Natural T cells, and notably the CD4+ subset, are related to atopy and total IgE levels (45). Also, the number of IFN-γ–producing CD8+ T cells is related to asthma severity, to bronchial hyperresponsiveness, and to blood eosinophilia (46). In some types of cancer, such as colorectal, esophageal, or gallbladder carcinoma, immunohistochemical identification of tumor-infiltrating CD8+ T lymphocytes has been shown to correlate with an improved overall survival (47, 48). In a single case report, the long-term survival of a 65-year-old man who underwent pancreaticoduodenectomy with portal vein resection for pancreatic cancer has been attributed to the response of CD8+ T cells to the cancer (49). This finding was corroborated by a study based on tumor specimens obtained from 80 patients with pancreatic adenocarcinomas, which showed that CD4/8+/+ status was an independent favorable prognostic factor after surgical treatment (50). Using an animal model, Karagiannis et al. (51) established that IgE antibodies commonly involved in allergic responses could trigger an immune response against ovarian cancer. In their experiment, injection of tumor-bearing mice with peripheral blood mononuclear cells and MOv18 IgE led to infiltration of monocytes into the tumors and prolonged survival of the mice, providing evidence that tumor-specific IgE antibodies may be exploited for immunotherapy of cancer.

Immune Surveillance of Cancer and the Pancreas

The concept of immune surveillance and editing stresses the importance of the immune system in eliminating preneoplastic cells and thus safeguarding the body against cancer through an IFN-γ–dependent mechanism. The immune cells that have been implicated in surveillance are natural killer (NK) cells, NK-T cells, CTLs, and γδ T cells. These cells come from both the innate (NK and NK-T cells) and adaptive, antigen-specific (γδ, αβ T cells) immune system. All of these cells have the potential to survey the pancreas; most of these cells are active during allergic responses.

During transformation, preneoplastic cells may lose MHC expression or express potentially immunogenic tumor antigens. Loss of MHC expression could lead to recognition by the innate immune system, whereas expression of potential tumor-specific antigens could lead to recognition by T cells. Under conditions of stress (i.e., allograft rejection, inflammation, or neoplastic transformation), the pancreas has been shown to up-regulate the MHC-like molecules MIC-A and MIC-B (52-54). MIC-A and MIC-B act through the NKG2D costimulatory molecule and directly activate or costimulate NK cells, γδ T cells, CD8+ T cells, and NK-T cells, thus allowing for surveillance of the pancreas by the innate and adaptive immune system (55).

A case for a link between allergy and immune surveillance could be made for the cell types mentioned above. Cells of the innate immune system (NK and NK-T cells) would be expected to be part of the immune surveillance process. Recent conflicting data has been presented on whether NK-T cells are a part of the immune surveillance process in relation to allergy. Allergy is mediated by type 2 responses, which are characterized by the cytokines interleukin (IL)-4 and IL-10. Conflicting data from Oishi (56) and Saikai (57) have shown that during allergic responses, the number of NK-T cells is reduced, or increases over time during constant exposure to allergen, respectively, thus creating a possible link between NK-T cells in allergy and immune surveillance.

A link between NK cell immune surveillance and allergy would be unexpected because the predominant cytokine milieu during allergic responses (IL-4, IL-13) does not correspond to what is expected during immune editing (IFN-γ). Recent evidence, however, suggests that allergy, specifically asthma, may not be tipped so heavily toward a type 2 (IL-4, IL-13) response. Kuepper et al. (58) have shown increased type 1 CTL activity (IFN-γ) in asthmatics that depends on the activation and cell-to-cell contact with NK cells, suggesting that NK cells may be active during an allergic response.

γδ T cells may also be a bridge between immune surveillance and allergy. γδ T cells are a subset of T cells that are not MHC-restricted, have varied receptor diversity, and an unknown antigenic target(s). They can be activated by MIC-A protein expression making them more like innate effectors, but they have many proposed roles, including tissue repair, tumor rejection, and regulation of inflammation (59). They have been shown to be potent IFN-γ secretors and should counteract type 2 allergic responses. In fact, a lack of γδ T cells has been correlated with increased contact hypersensitivity in the skin (60). However, many groups have shown that γδ T cells in asthma are increased and show increased IL-4 and tumor necrosis factor secretion. Because they are essential for IgE and eosinophil infiltration in allergic asthma airway inflammation (6163) and because of their increase and ability to recognize the MIC-A/B proteins, γδ T cells may connect allergy and immunosurveillance.

The cells of the adaptive immune response, α/β T cells, are also involved in immune surveillance and could cross-over from allergic responses. Antigen-specific T helper cells and CTLs are thought to be important in immune surveillance and are traditionally associated with type 1 responses that secrete IL-2 and IFN-γ. Dutton et al. (64, 65), however, have shown the effectiveness of type 2 CTL cells in antitumor response. These CTLs are as lytic as traditional IFN-γ–secreting CTLs, but secrete and respond to IL-4. As stated above, classic IFN-γ–secreting CTLs have been found in asthmatics that rely on the activation and cell-to-cell contact with NK cells. Recent data in murine and human studies have shown the existence of T helper 1 responses (IFN-γ secreting) contributing to airway inflammation in asthmatics (66-68). These data suggest that the hypersensitivity found in allergic responses may lead to a broader activation of the immune system and to increased immune surveillance against tumors.

A Possible Link between Allergy and Tumor Immunotherapy?

Although many tumor immunotherapies have focused on type 1 responses, some early murine tumor models using IL-4–secreting tumor vaccines showed success and one recent study suggests that IL-4 may enhance type 1 responses possibly by acting on dendritic cells (69-71). Evidence for a possible link between allergy and tumor immunotherapy of pancreatic cancer was seen in studies designed to enhance the activation and maturation of dendritic cells. Pancreatic cancer cells engineered to secrete granulocyte macrophage colony-stimulating factor, a potent dendritic cell activator, were used as a vaccine in a clinical trial against minimal residual disease. Five of 14 patients receiving the vaccine showed a significant increase in eosinophils both systemically and at the vaccine site. Three of these patients went on to have prolonged long-term survival (>39 months; ref. 72). In addition, one of these three patients had experienced multiple recurring systemic rashes and recall responses at old vaccine sites that seem to be mediated by eosinophils and T cells. Given the predominance of eosinophils in classic allergic reaction, these data suggest a link between the mediators of allergy and the classic type 1 antitumor responses.

In conclusion, although this meta-analysis is based on studies that were not all designed to address appropriately this association, its results provide some evidence that, unlike other exposure variables such as smoking or pancreatitis, which increase the risk of pancreatic cancer, allergies, particularly atopic allergy, may protect against pancreatic cancer. The notion that the immune system itself could regulate cancer development through a functional cancer immunosurveillance process has led to the development of monoclonal antibody therapies and cancer vaccines. However, current knowledge is insufficient to suggest a practical way to immunize high-risk patients against pancreatic cancer, but there is sufficient information to justify continued efforts to stimulate the immune system as a therapeutic measure in the treatment of pancreatic cancer.

Grant support: CD Smithers Foundation, Solvay Pharmaceuticals Corporation, and the Italian Association for Cancer Research.

The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

1
Jemal A, Murray T, Samuels A, Ghafoor A, Ward E, Thun MJ. Cancer statistics, 2003.
CA Cancer J Clin
2003
;
53
:
5
–26.
2
Lowenfels AB, Maisonneuve P. Epidemiology and prevention of pancreatic cancer.
Jpn J Clin Oncol
2004
;
34
:
238
–44.
3
Vena JE, Bona JR, Byers TE, Middleton EJ, Swanson MK, Graham S. Allergy-related diseases and cancer: an inverse association.
Am J Epidemiol
1985
;
122
:
66
–74.
4
Kallen B, Gunnarskog J, Conradson TB. Cancer risk in asthmatic subjects selected from hospital discharge registry.
Eur Respir J
1993
;
6
:
694
–7.
5
Brenner AV, Linet MS, Fine HA, et al. History of allergies and autoimmune diseases and risk of brain tumors in adults.
Int J Cancer
2002
;
99
:
252
–9.
6
Wiemels JL, Wiencke JK, Sison JD, Miike R, McMillan A, Wrensch M. History of allergies among adults with glioma and controls.
Int J Cancer
2002
;
98
:
609
–15.
7
Bosetti C, Talamini R, Franceschi S, Negri E, Giacosa A, La Vecchia C. Allergy and the risk of selected digestive and laryngeal neoplasms.
Eur J Cancer Prev
2004
;
13
:
173
–6.
8
Severson RK, Davis S, Thomas DB, Stevens RG, Heuser L, Sever LE. Acute myelocytic leukemia and prior allergies.
J Clin Epidemiol
1989
;
42
:
995
–1001.
9
Wen W, Shu XO, Linet MS, et al. Allergic disorders and the risk of childhood acute lymphoblastic leukemia (United States).
Cancer Causes Control
2000
;
11
:
303
–7.
10
Schuz J, Morgan G, Bohler E, Kaatsch P, Michaelis J. Atopic disease and childhood acute lymphoblastic leukemia.
Int J Cancer
2003
;
105
:
255
–60.
11
Grulich AE, Vajdic CM, Kaldor JM, et al. Birth order, atopy, and risk of non-Hodgkin lymphoma.
J Natl Cancer Inst
2005
;
97
:
587
–94.
12
Hedderson MM, Malone KE, Daling JR, White E. Allergy and risk of breast cancer among young women (United States).
Cancer Causes Control
2003
;
14
:
619
–26.
13
Haynes RB, Wilczynski N, McKibbon KA, Walker CJ, Sinclair JC. Developing optimal search strategies for detecting clinically sound studies in MEDLINE.
J Am Med Inform Assoc
1994
;
1
:
447
–58.
14
Shojania KG, Bero LA. Taking advantage of the explosion of systematic reviews: an efficient MEDLINE search strategy.
Eff Clin Pract
2001
;
4
:
157
–62.
15
Dickersin K, Scherer R, Lefebvre C. Identifying relevant studies for systematic reviews.
BMJ
1994
;
309
:
1286
–91.
16
Greenland S. Quantitative methods in the review of epidemiologic literature.
Epidemiol Rev
1987
;
9
:
1
–30.
17
Sterne JA, Juni P, Schulz KF, Altman DG, Bartlett C, Egger M. Statistical methods for assessing the influence of study characteristics on treatment effects in “meta-epidemiological” research.
Stat Med
2002
;
21
:
1513
–24.
18
van Houwelingen HC, Arends LR, Stijnen T. Advanced methods in meta-analysis: multivariate approach and meta-regression.
Stat Med
2002
;
21
:
589
–624.
19
DerSimonian R, Laird N. Meta-analysis in clinical trials.
Control Clin Trials
1986
;
7
:
177
–88.
20
Copas JB, Shi JQ. A sensitivity analysis for publication bias in systematic reviews.
Stat Methods Med Res
2001
;
10
:
251
–5.
21
Macaskill P, Walter SD, Irwig L. A comparison of methods to detect publication bias in meta-analysis.
Stat Med
2001
;
20
:
641
–54.
22
Lin RS, Kessler II. A multifactorial model for pancreatic cancer in man. Epidemiologic evidence.
JAMA
1981
;
245
:
147
–52.
23
Gold EB, Gordis L, Diener MD, et al. Diet and other risk factors for cancer of the pancreas.
Cancer
1985
;
55
:
460
–7.
24
Mack TM, Yu MC, Hanisch R, Henderson BE. Pancreas cancer and smoking, beverage consumption, and past medical history.
J Natl Cancer Inst
1986
;
76
:
49
–60.
25
McWhorter WP. Allergy and risk of cancer. A prospective study using NHANESI followup data.
Cancer
1988
;
62
:
451
–5.
26
Mills PK, Beeson WL, Abbey DE, Fraser GE, Phillips RL. Dietary habits and past medical history as related to fatal pancreas cancer risk among Adventists.
Cancer
1988
;
61
:
2578
–85.
27
Farrow DC, Davis S. Risk of pancreatic cancer in relation to medical history and the use of tobacco, alcohol and coffee.
Int J Cancer
1990
;
45
:
816
–20.
28
La Vecchia C, Negri E, D'Avanzo B, et al. Medical history, diet and pancreatic cancer.
Oncology
1990
;
47
:
463
–6.
29
Jain M, Howe GR, St Louis P, Miller AB. Coffee and alcohol as determinants of risk of pancreas cancer: a case-control study from Toronto.
Int J Cancer
1991
;
47
:
384
–9.
30
Bueno dMH, Maisonneuve P, Moerman CJ, Walker AM. Aspects of medical history and exocrine carcinoma of the pancreas: a population-based case-control study in the Netherlands.
Int J Cancer
1992
;
52
:
17
–23.
31
Kalapothaki V, Tzonou A, Hsieh CC, Toupadaki N, Karakatsani A, Trichopoulos D. Tobacco, ethanol, coffee, pancreatitis, diabetes mellitus, and cholelithiasis as risk factors for pancreatic carcinoma.
Cancer Causes Control
1993
;
4
:
375
–82.
32
Dai Q, Zheng W, Ji BT, et al. Prior immunity-related medical conditions and pancreatic-cancer risk in Shanghai.
Int J Cancer
1995
;
63
:
337
–40.
33
Eriksson NE, Holmen A, Hogstedt B, Mikoczy Z, Hagmar L. A prospective study of cancer incidence in a cohort examined for allergy.
Allergy
1995
;
50
:
718
–22.
34
Silverman DT, Schiffman M, Everhart J, et al. Diabetes mellitus, other medical conditions and familial history of cancer as risk factors for pancreatic cancer.
Br J Cancer
1999
;
80
:
1830
–7.
35
Stolzenber-Solomon RZ, Pietinen P, Taylor PR, Virtamo J, Albanes D. A prospective study of medical conditions, anthropometry, physical activity, and pancreatic cancer in male smokers (Finland).
Cancer Causes Control
2002
;
13
:
417
–26.
36
Holly EA, Eberle CA, Bracci PM. Prior history of allergies and pancreatic cancer in the San Francisco Bay area.
Am J Epidemiol
2003
;
158
:
432
–41.
37
Fisherman EW. Does the allergic diathesis influence malignancy?
J Allergy
1960
;
31
:
74
–8.
38
Briggs NC, Levine RS, Brann EA. Allergies and risk of non-Hodgkin's lymphoma by subtype.
Cancer Epidemiol Biomarkers Prev
2002
;
11
:
401
–7.
39
McKee WD, Arnold CA, Perlman MD. A double-blind study of the comparative incidence of malignancy and allergy.
J Allergy
1967
;
39
:
294
–301.
40
Hughes WF, Raitz RL. A comparison of cancer occurrence in allergic and nonallergic populations.
Ann Allergy
1979
;
43
:
163
–4.
41
Lindelof B, Sigurgeirsson B, Wahlgren CF, Eklund G. Chronic urticaria and cancer: an epidemiological study of 1155 patients.
Br J Dermatol
1990
;
123
:
453
–6.
42
Spector L, Groves F, DeStefano F, et al. Medically recorded allergies and the risk of childhood acute lymphoblastic leukaemia.
Eur J Cancer
2004
;
40
:
579
–84.
43
Mills PK, Beeson WL, Fraser GE, Phillips RL. Allergy and cancer: organ site-specific results from the Adventist Health Study.
Am J Epidemiol
1992
;
136
:
287
–95.
44
Pompei R, Lampis G, Ingianni A, Nonnis D, Ionta MT, Massidda B. Allergy and tumour outcome after primary cancer therapy.
Int Arch Allergy Immunol
2004
;
133
:
174
–8.
45
Magnan A, Mely L, Prato S, et al. Relationships between natural T cells, atopy, IgE levels, and IL-4 production.
Allergy
2000
;
55
:
286
–90.
46
Magnan AO, Mely LG, Camilla CA, et al. Assessment of the Th1/Th2 paradigm in whole blood in atopy and asthma. Increased IFN-γ-producing CD8(+) T cells in asthma.
Am J Respir Crit Care Med
2000
;
161
:
1790
–6.
47
Naito Y, Saito K, Shiiba K, et al. CD8+ T cells infiltrated within cancer cell nests as a prognostic factor in human colorectal cancer.
Cancer Res
1998
;
58
:
3491
–4.
48
Schumacher K, Haensch W, Roefzaad C, Schlag PM. Prognostic significance of activated CD8(+) T cell infiltrations within esophageal carcinomas.
Cancer Res
2001
;
61
:
3932
–6.
49
Abe M, Kondo S, Hirano S, et al. Long-term survival after radical resection of advanced pancreatic cancer: a case report with special reference to CD8+ T-cell infiltration.
Int J Gastrointest Cancer
1904
;
33
:
107
–10.
50
Fukunaga A, Miyamoto M, Cho Y, et al. CD8+ tumor-infiltrating lymphocytes together with CD4+ tumor-infiltrating lymphocytes and dendritic cells improve the prognosis of patients with pancreatic adenocarcinoma.
Pancreas
2004
;
28
:
26
–e31.
51
Karagiannis SN, Wang Q, East N, et al. Activity of human monocytes in IgE antibody-dependent surveillance and killing of ovarian tumor cells.
Eur J Immunol
2003
;
33
:
1030
–40.
52
Hankey KG, Drachenberg CB, Papadimitriou JC, et al. MIC expression in renal and pancreatic allografts.
Transplantation
2002
;
73
:
304
–6.
53
Groh V, Rhinehart R, Secrist H, Bauer S, Grabstein KH, Spies T. Broad tumor-associated expression and recognition by tumor-derived γδ T cells of MICA and MICB.
Proc Natl Acad Sci U S A
1999
;
96
:
6879
–84.
54
Groh V, Bahram S, Bauer S, Herman A, Beauchamp M, Spies T. Cell stress-regulated human major histocompatibility complex class I gene expressed in gastrointestinal epithelium.
Proc Natl Acad Sci U S A
1996
;
93
:
12445
–50.
55
Ogasawara K, Hamerman JA, Ehrlich LR, et al. NKG2D blockade prevents autoimmune diabetes in NOD mice.
Immunity
2004
;
20
:
757
–67.
56
Oishi Y, Sakamoto A, Kurasawa K, et al. CD4CD8 T cells bearing invariant Vα24JαQ TCR α-chain are decreased in patients with atopic diseases.
Clin Exp Immunol
2000
;
119
:
404
–11.
57
Saikai T, Tanaka H, Sato N, Abe S, Matsuura A. Mushroom plant workers experience a shift towards a T helper type 2 dominant state: contribution of innate immunity to spore antigen.
Clin Exp Immunol
2004
;
135
:
119
–24.
58
Kuepper M, Koester K, Bratke K, et al. Increase in Ksp37-positive peripheral blood lymphocytes in mild extrinsic asthma.
Clin Exp Immunol
2004
;
137
:
359
–65.
59
Jameson J, Witherden D, Havran WL. T-cell effector mechanisms: γδ and CD1d-restricted subsets.
Curr Opin Immunol
2003
;
15
:
349
–53.
60
Guan H, Zu G, Slater M, Elmets C, Xu H. γδT cells regulate the development of hapten-specific CD8+ effector T cells in contact hypersensitivity responses.
J Invest Dermatol
2002
;
119
:
137
–42.
61
Wisnewski AV, Herrick CA, Liu Q, Chen L, Bottomly K, Redlich CA. Human γ/δ T-cell proliferation and IFN-γ production induced by hexamethylene diisocyanate.
J Allergy Clin Immunol
2003
;
112
:
538
–46.
62
Hamzaoui A, Kahan A, Ayed K, Hamzaoui K. T cells expressing the γδ receptor are essential for Th2-mediated inflammation in patients with acute exacerbation of asthma.
Mediators Inflamm
2002
;
11
:
113
–9.
63
Svensson L, Lilliehook B, Larsson R, Bucht A. γδ T cells contribute to the systemic immunoglobulin E response and local B-cell reactivity in allergic eosinophilic airway inflammation.
Immunology
2003
;
108
:
98
–108.
64
Croft M, Carter L, Swain SL, Dutton RW. Generation of polarized antigen-specific CD8 effector populations: reciprocal action of interleukin (IL)-4 and IL-12 in promoting type 2 versus type 1 cytokine profiles.
J Exp Med
1994
;
180
:
1715
–28.
65
Dobrzanski MJ, Reome JB, Hollenbaugh JA, Dutton RW. Tc1 and Tc2 effector cell therapy elicit long-term tumor immunity by contrasting mechanisms that result in complementary endogenous type 1 antitumor responses.
J Immunol
2004
;
172
:
1380
–90.
66
Boniface S, Koscher V, Mamessier E, et al. Assessment of T lymphocyte cytokine production in induced sputum from asthmatics: a flow cytometry study.
Clin Exp Allergy
2003
;
33
:
1238
–43.
67
Hansen G, Berry G, DeKruyff RH, Umetsu DT. Allergen-specific Th1 cells fail to counterbalance Th2 cell-induced airway hyperreactivity but cause severe airway inflammation.
J Clin Invest
1999
;
103
:
175
–83.
68
Randolph DA, Carruthers CJ, Szabo SJ, Murphy KM, Chaplin DD. Modulation of airway inflammation by passive transfer of allergen-specific Th1 and Th2 cells in a mouse model of asthma.
J Immunol
1999
;
162
:
2375
–83.
69
Golumbek PT, Lazenby AJ, Levitsky HI, et al. Treatment of established renal cancer by tumor cells engineered to secrete interleukin-4.
Science
1991
;
254
:
713
–6.
70
Cayeux S, Richter G, Noffz G, Dorken B, Blankenstein T. Influence of gene-modified (IL-7, IL-4, and B7) tumor cell vaccines on tumor antigen presentation.
J Immunol
1997
;
158
:
2834
–41.
71
Schuler T, Kammertoens T, Preiss S, Debs P, Noben-Trauth N, Blankenstein T. Generation of tumor-associated cytotoxic T lymphocytes requires interleukin 4 from CD8(+) T cells.
J Exp Med
2001
;
194
:
1767
–75.
72
Jaffee EM, Hruban RH, Biedrzycki B, et al. Novel allogeneic granulocyte-macrophage colony-stimulating factor-secreting tumor vaccine for pancreatic cancer: a phase I trial of safety and immune activation.
J Clin Oncol
2001
;
19
:
145
–56.