Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2014 Feb 4.
Published in final edited form as: Breast Cancer Res Treat. 2013 Apr 30;139(2):477–488. doi: 10.1007/s10549-013-2549-3

Pretreatment levels of circulating Th1 and Th2 cytokines, and their ratios, are associated with ER-negative and triple negative breast cancers

Chi-Chen Hong 1,, Song Yao 2, Susan E McCann 3, Ree Y Dolnick 4, Paul K Wallace 5, Zhihong Gong 6, Lei Quan 7, Kelvin P Lee 8, Sharon S Evans 9, Elizabeth A Repasky 10, Stephen B Edge 11, Christine B Ambrosone 12
PMCID: PMC3912696  NIHMSID: NIHMS541593  PMID: 23624818

Abstract

Immune signatures in breast tumors differ by estrogen receptor (ER) status. The purpose of this study was to assess associations between ER phenotypes and circulating levels of cytokines that co-ordinate cell-mediated [T-helper type 1 (Th1)] and humoral [T-helper type 2 (Th2)] immunity. We conducted a case–case comparison of 523 women with newly diagnosed breast cancer to evaluate associations between 27 circulating cytokines, measured using Luminex XMap technology, and breast cancer phenotypes [ER vs. ER+; triple negative breast cancer (TNBC) vs. luminal A (LumA)]. Ratios of Th1 to Th2 cytokines were also evaluated. Levels of interleukin (IL)-5, a Th-2 cytokine, were higher in ER than in ER+ tumors. The highest tertile of IL-5 was more strongly associated with ER (OR = 2.33, 95 % CI 1.40–3.90) and TNBCs (OR = 2.78, 95 % CI 1.53–5.06) compared to ER+ and LumA cancers, respectively, particularly among premenopausal women (OR = 4.17, 95 % CI 1.86–9.34, ER vs. ER+; OR = 5.60, 95 % CI 2.09–15.01, TNBC vs. LumA). Elevated Th1 cytokines were also detected in women with ER and TNBCs, with women in the highest tertile of interferon α2 (OR = 2.39, 95 % CI 1.31–4.35) or tumor necrosis factor-α (OR = 2.27, 95 % CI 1.21–4.26) being twice as likely to have TNBC versus LumA cancer. When cytokine ratios were examined, women with the highest ratios of Th1 cytokines to IL-5 levels were least likely to have ER or TNBCs compared to ER+ or LumA cancers, respectively. The strongest associations were in premenopausal women, who were up to 80 % less likely to have TNBC than LumA cancers (IL-12p40/IL-5, OR = 0.19, 95 % CI 0.07–0.56). These findings indicate that immune function is associated with ER and TNBC and may be most relevant among younger women, who are likely to be diagnosed with these aggressive phenotypes.

Keywords: Estrogen negative breast cancer, Triple negative breast cancer, Cytokines Immune function

Introduction

Breast cancer intrinsic subtypes, now characterized by immunohistochemistry (IHC), have important differences in prognosis, with LumA (ER+ and/or PR+ and HER2) having better prognosis and basal-like tumors (ER, PR, HER2, CK5/6+ and/or EGFR+) the poorest [1, 2]. Most breast cancers with a triple negative phenotype (ER, PR, HER2), i.e., 70–90 %, are also basal-like [35]. Younger women, BRCA1 carriers, and women of African or Hispanic ancestry have higher rates of basal-like and TNBC [6, 7]. Although subtype-specific risk factors have been understudied, there do appear to be differences in some reproductive and hormonal factors for ER+ and TNBC [6, 8, 9].

Pro-inflammatory biomarkers and immune-response may be related to breast cancer risk and/or prognosis [10, 11]. For ER and TNBC, gene expression studies show that inflammation and immune-related signatures are quantitatively and qualitatively different between ER+ and ER cancers, and important for disease prognosis [1214]. Therefore, we hypothesized that cytokines functioning as immune system signaling chemical messengers could be related to ER and TNBC.

Cytokines can be classified into broad categories that include T-helper type 1 (Th1) and T-helper type 2 (Th2), which are generally antagonistic to each other (see Fig. 1). Th1 adaptive immunity, important for mounting an effective anti-tumor immune response [15, 16], is characterized by IFNγ, TNFα, IL-12, GM-CSF, and CXCL10 (IP10), and can activate macrophages toward a M1 phenotype, further promoting a Th1 response. Th-2 cytokines include IL-4, IL-5, IL-10, CCL2, CCL7, and CCL11, and are involved in allergy and M2 polarization of macrophages, which are important in mediating humoral immunity and appear to promote mammary tumor progression [15, 17]. Several cytokines associated with chronic inflammation, such as CXCL8 and IL-6 [18, 19], have been linked to poorer breast cancer survival [20], particularly for TNBC [13]. Previous research examining associations between cytokines and ER and TNBCs have mostly focused on tumor-associated changes in the microenvironment. Here we investigate potential associations with circulating cytokine levels involved in Th-1 and Th-2 immunity and chronic inflammation, which may provide greater understanding into the role of constitutional host immunity on breast cancer subtypes. In addition, we also examined several cytokine ratios to reflect the balance between Th1 and Th2 cell types since these cell subsets are often inversely regulated and their balance can be influenced by many factors [2123].

Fig. 1.

Fig. 1

Role of cytokines in Th1 and Th2 differentiation of lymphocytes. CD4+ T-helper lymphocytes are able to augment both cellular and humoral immune responses. During an immune response, cytokines as a component of the microenvironment present during T-cell priming can influence the developmental pathway taken by responding naïve CD4+ T cells. The Th1 developmental pathway is largely driven by IL-12 activation, producing high levels of IFNγ and TNFα upon antigen stimulation, and is responsible for regulating cell-mediated immunity. The Th1 cytokine IFNγ also polarizes macrophages (innate immune cells) towards the M1 phenotype. Th2 differentiation, in contrast, is largely driven by IL-4 and is characterized by IL-4, IL-5, IL-10, and IL-13 secretion, which are primarily involved in the coordination of humoral immunity, eosinophilic inflammation, and control of helminth infections. The Th2 cytokines IL-4 and IL-13, as well as CCL2 from monocytes, drives macrophages toward M2 polarization leading to high expression of IL-10 and IL1RA. The Th1 and Th2 pathways are regulated by a balance of positive and antagonistic feedback loops. IFNγ enhances further Th1 development and suppresses Th2 differentiation, while IL-4 and IL-10 supports Th2 differentiation and suppresses Th1 differentiation. In addition, CD4+ CD25+ Foxp3+ T-regulatory cells (Tregs), a subset of CD4+ T cells, mediate general immune suppression and can drive macrophages toward “M2-like” phenotypes, which are important for the prevention of autoimmunity, but can suppress anti-tumor immunity [17, 80, 106, 107]

Patients and methods

Study population and blood collection

Data and plasma samples from 523 non-Hispanic Caucasian women with incident invasive breast cancer treated at Roswell Park Cancer Institute (RPCI) from December 2003 to June 2010 were obtained from the Institute’s Data Bank and Biorepository (DBBR) [24, 25]. Prior to receipt of treatment (including surgery), newly diagnosed patients consent to DBBR, to provide non-fasting blood samples, complete an epidemiological questionnaire, and permit linkage of samples to clinical information. Blood samples are drawn in phlebotomy and transported via a pneumatic tube system to the laboratory for processing, where they are centrifuged and automatically aliquoted into chemically inert plastic straws. The time from blood draw to storage in liquid nitrogen is maintained at 1 h of draw to minimize variable analyte degradation. For most participants in our study, samples were obtained prior to surgery (n = 475, 91 %) and adjuvant therapy (n = 519, 99 %). This research was approved by the Institutional Review Board (IRB) of RPCI.

Clinical information

Samples were linked with clinical data derived from a prospective database of the RPCI Breast Program by the RPCI Clinical Data Network (CDN). Molecular subtypes were defined based on ER, PR, and HER2 status determined routinely by IHC or FISH in the Department of Pathology and classified as LumA (ER+ and/or PR+, HER2) and triple negative (ER, PR, and HER2). Because IHC staining of CK 5/6 or EGFR are not routinely performed in pathology at RPCI, we could not distinguish basal-like and unclassified subtypes, and both were included in the triple negative group.

Postmenopausal status was defined as 12 consecutive months of amenorrhea and/or a bilateral salpingo-oophorectomy as reported in a self-administered questionnaire. For women without self-reported menopausal status, data were abstracted from patient medical records. Where medical record data were unavailable (n = 9), we used an age cut point, with premenopausal status defined as 56 years of age or younger.

Luminex xMAP (Multi-Analyte Profiling) immune-bead array assays

Prior to these analyses, we conducted a pilot study with a series of QC experiments to investigate the effects of various sample processing and storage procedures on the recovery of cytokines and growth factors, as well as to compare across commercially available Luminex® bead-based cytokine kits to identify those with the best reproducibility and sensitivity.

In the analyses reported herein, plasma samples were used to measure a panel of 27 cytokines. Five analytes were multiplexed (IL-1β, IL-4, IL-6, IL-10, GM-CSF) and assayed using high sensitivity kits (Millipore, HSCYT-MAG-60SK), and 22 analytes (IFNγ, IFNα2, TNFα,TNFβ, IL12p40, IL12p70, IL1α, IL1RA, IL2, IL5, IL15, GCSF, CCL2, CCL3, CCL4, CCL7, CCL11, CCL22, CXCL1, CXCL8, CXCL10, CX3CL1) were assayed using regular kits (Millipore, HCYTOMAG-60 K). For standard curves, reconstituted top standards (high sensitivity kits, 2000 pg/ml; cytokine-chemokine kits, 10000 pg/ml) were serially diluted 1:3 with assay buffer for nine point curves. Assays were assembled using 96-well plates. Each plate contained samples (in duplicates), standards, and internal quality controls (QCs 1 and 2). Analyte capture was carried out according to manufacturer’s instructions (Millipore, Cat # 40–285). Data were acquired using Luminex 100 with xPONENT version 3.1 software, and concentrations measured using BeadView Analysis Software. Plate-specific standard curves were generated using the “Best Fit” curve fitting routine which automatically selects the best curve algorithm for each analyte. When necessary, obvious outliers were omitted prior to generating standard curves. Intra-plate coefficient of variations (CVs) ranged from 1.4 to 7.5 %. Interplate CVs of 30 non-blinded company-supplied control samples varied between 2.7 and 11.9 %, with only four analytes having CVs greater than 10 % (IL-4, IL-6, IL-10, CX3CL1). The CVs observed in our study were only slightly higher than those observed with conventional ELISA assays. Thus, we are confident in the quality of data generated using this method.

Statistical analysis

Cytokines were categorized into tertiles corresponding to ‘low, medium, and high’. The proportion of non-detectable values exceeded one-third for 5 cytokines (TNFβ, IL-1α, IL-1RA, IL-2, IL-15), which were categorized as “low”, with remaining samples dichotomized at the median for “medium” and “high” levels. Because different subsets of T helper cells regulate each other, often inversely, we calculated several cytokine ratios, primarily to reflect the balance between Th1 and Th2 cell types. If markers were below the detectable assay limit, the median value between zero and the reliable lower limit was assigned to enable these calculations. Specific cutpoints and all ratios examined are shown in Online Resource Table 1, along with detectable assay limits, interplate coefficient of variations for each cytokine measured, and proportion of samples with non-detectable cytokine levels.

Odds ratios (OR) and 95 % confidence intervals (CIs) for associations between cytokine measures and ER versus ER+ tumors and triple negative versus LumA cancers were estimated with unconditional logistic regression. Since ER and TNBCs are more prevalent in younger women, we also stratified by menopausal status. To test for linear trend, categorical cytokine levels were assessed in models as a continuous ordinal variable. To test multiplicative interactions by menopausal status, cross-product terms between menopausal status and category-specific log transformed median cytokine levels were included in multivariate models. Using the Wald χ2 test, statistical significance indicated a difference in slopes associated with cytokine levels between pre- and postmenopausal women. All analyses were further adjusted for AJCC breast cancer stage, histological grade, age and season and timing of blood draw in relation to receipt of treatment (see Online Resource Table 1).

All tests for significance were 2-sided and statistical analyses were conducted using SAS for Windows, Version 9.3, and a false discovery rate (FDR)-adjusted p value (q) was used to correct for multiple comparisons [26]. The significance level of individual cytokines was corrected for a total of 27 tests, and cytokine ratios were corrected for a total of 37 tests.

Results

Approximately two-thirds of the women were older than 50 years (Table 1), and the majority had stage I breast cancer; 29 % had ER breast cancer, and 22 % had TNBCs. As expected, ER and TNBCs were of higher AJCC stage, histological grade, and size compared to ER+ and LumA cancers, respectively.

Table 1.

Participant characteristics of 523 women with invasive breast cancer

Characteristics All women ER status
Subtype
ER+
(n = 370)
ER
(n = 153)
p LumA
(n = 332)
TNBC
(n = 116)
p
Age at diagnosis, years, mean ± SD, n (%) 56.8 ± 12.9 57.6 ± 12.8 55.0 ± 13.2 0.03 57.6 ± 12.7 56.8 ± 12.9 0.53
  <50 years 172 (32.9) 118 (31.9) 54 (35.3) 0.12 105 (31.6) 35 (30.2) 0.52
  50–64 years 202 (38.6) 137 (37.0) 65 (42.5) 124 (37.4) 50 (43.1)
  ≥65 years 149 (28.5) 115 (31.1) 34 (22.2) 103 (31.0) 31 (26.7)
Body mass index, kg/m2, mean ± SDa 28.2 ± 6.4 28.4 ± 6.5 27.6 ± 6.2 0.24 28.5 ± 6.6 27.9 ±6.1 0.40
  <25 156 (29.8) 111 (30.0) 45 (29.4) 0.23 99 (29.8) 32 (27.6) 0.12
  25–<30 173 (33.1) 118 (31.9) 55 (36.0) 103 (31.0) 40 (34.5)
  ≥30 174 (33.3) 130 (35.1) 44 (28.8) 120 (36.1) 35 (30.2)
  Unknown 20 (3.8) 11 (3.0) 9 (5.9) 10 (3.0) 9 (7.8)
Menopausal status, n (%) 0.78 0.82
  Premenopausal 207 (39.6) 145 (39.2) 62 (40.5) 127 (38.3) 43 (37.1)
  Postmenopausal 316 (60.4) 225 (60.8) 91 (59.5) 205 (61.8) 73 (62.9)
Stage, n (%) 0.002 0.002
  I 304 (58.1) 236 (63.8) 68 (44.4) 217 (65.4) 55 (47.4)
  IIA 119 (22.8) 77 (20.8) 42 (27.5) 66 (19.9) 32 (27.6)
  IIB 43 (8.2) 23 (6.2) 20 (13.1) 18 (5.4) 16 (13.8)
  IIIA 34 (6.5) 21 (5.7) 13 (8.5) 20 (6.0) 6 (5.2)
  IIIB/IIIC/IV 21 (4.0) 12 (3.2) 9 (5.9) 11 (3.3) 6 (5.2)
  TX 2 (0.4) 1 (0.3) 1 (0.7) 1 (0.9)
Histological grade, n (%) <0.0001 <0.0001
  I 43 (8.2) 42 (11.3) 1 (0.7) 40 (12.1) 1 (0.9)
  II 132 (25.2) 114 (30.8) 18 (11.8) 105 (31.6) 14 (12.1)
  III 341 (65.2) 210 (56.8) 131 (85.6) 185 (55.7) 99 (85.3)
  Unknown 7 (1.3) 4 (1.1) 3 (2.0) 2 (0.6) 2 (1.7)
Number of positive nodes 0.02 0.12
  0 361 (69.0) 262 (70.8) 99 (64.7) 236 (71.1) 83 (71.5)
  1–2 88 (16.8) 62 (16.8) 26 (17.0) 53 (16.0) 19 (16.4)
  ≥3 57 (10.9) 31 (8.4) 26 (17.0) 29 (8.7) 14 (12.1)
  Unknown 17 (3.3) 15 (4.1) 2 (1.3) 14 (4.2) 0 (0)
Tumor size, cm, mean ± SDb 1.7 ± 1.3 1.6 ± 1.2 2.0 ±1.6 0.003 1.6 ± 1.2 2.0 ± 1.4 0.006
  <1 cm 122 (23.3) 90 (24.3) 32 (20.9) 0.002 83 (25.0) 22 (19.0) 0.0002
  1–<2 cm 263 (50.3) 200 (54.0) 63 (41.2) 184 (55.4) 50 (43.1)
  ≥2 cm 126 (24.1) 74 (20.0) 52 (34.0) 62 (18.7) 38 (32.8)
  Unknown 12 (2.3) 6 (1.6) 6 (3.9) 3 (0.9) 6 (5.2)
a

Excludes 20 women with missing BMI values

b

Excludes 12 women with unknown tumor size

Association with ER and TNBCs

As shown in Figs. 2 and 3, both ER and TNBCs were associated with high levels of circulating IL-5, a Th2 cytokine. Overall, women in the highest versus lowest tertile had approximately 2.5-fold increased odds of ER breast cancer or TNBC compared to ER+ and LumA cancers, respectively. Associations were strongest among premenopausal women, with over fourfold increased odds of ER breast cancer (OR = 4.17, 95 % CI 1.86–9.34; p-interaction by menopausal status = 0.08) and 5.6-fold increased odds of TNBC (OR = 5.60, 95 % CI 2.09– 15.01; p-interaction by menopausal status = 0.12). Although ORs were above unity for postmenopausal women, none were significant. High levels of Th1-related cytokines, IFNα2 and TNFα, were also associated with increased odds of ER and TNBC. Compared to women in the lowest tertile, women in the highest tertile were approximately twice as likely to have ER versus ER+ cancers or TNBC versus LumA cancers, with stronger relationships again observed among premenopausal women and not significant among postmenopausal women. Premenopausal women with high IFNα2 levels, for instance, were >4 times more likely to be ER (OR = 4.11, 95 % CI 1.70–9.92) than ER+, with similar estimates for TNBC versus LumA (OR = 4.55, 95 % CI 1.58–12.34). Tests for heterogeneity by menopausal status showed a trend towards statistical significance (p-int ≤ 0.09). Complete data for all associations examined are shown in Online Resource Tables 2 and 3.

Fig. 2.

Fig. 2

Statistically significant associations between circulating immune markers and ER breast cancer among women with invasive breast cancer. Odds ratios and 95 % CI provided for tertile 2 (Inline graphic) and tertile 3 (Inline graphic) of cytokines or cytokine ratios compared to tertile 1 as the reference group. All logistic regression models were adjusted for age at diagnosis (continuous), season (Apr-Sept, Oct-Mar), date of blood draw (continuous), timing of blood draw in relation to surgery and treatment (No prior treatment, postsurgical with no adjuvant treatment or receipt of neoadjuvant or adjuvant treatment), stage (I, IIA, IIB, IIIa, IIIB/IIIC/IV), grade (I, II, III, unknown). Full results for all cytokines and ratios are provided in Online Resource Table 2. *FDR-adjusted p values remain significant or borderline significant (q-trend ≤ 0.07) after significance for cytokines are corrected for a total of 27 cytokine tests, and cytokine ratios are corrected for a total of 37 tests. Using the Wald χ2 test, a difference in slopes associated with cytokine levels (as the log-transformed category specific median value analyzed as a continuous variable) was observed between pre- and postmenopausal women (p-int ≤ 0.10)

Fig. 3.

Fig. 3

Statistically significant associations between circulating immune markers and risk of triple negative versus LumA breast cancer. Eight women with undetermined tumor subtype were excluded from analyses. Odds ratios and 95 % CI provided for tertile 2 (Inline graphic) and tertile 3 (Inline graphic) of cytokines or cytokine ratios compared to tertile 1 as the reference group. All logistic regression models were adjusted for age at diagnosis (continuous), season (Apr– Sept, Oct–Mar), date of blood draw (continuous), timing of blood draw in relation to surgery and treatment (No prior treatment, postsurgical with no adjuvant treatment or receipt of neoadjuvant or adjuvant treatment), stage (I, IIA, IIB, IIIa, IIIB/IIIC/IV), and tumor grade (I, II, III, unknown). Full results for all cytokines and ratios are provided in Online Resource Table 3. *FDR-adjusted p values remain significant or borderline significant (q-trend ≤ 0.07) after significance for cytokines are corrected for a total of 27 cytokine tests, and cytokine ratios are corrected for a total of 37 tests. Using the Wald χ2 test, a difference in slopes associated with cytokine levels (as the log-transformed category specific median value analyzed as a continuous variable) was observed between pre- and postmenopausal women (p-int ≤ 0.10)

For several cytokines, associations were limited to either pre- or postmenopausal women. Among premenopausal women, high levels of CX3CL1 were associated with increased likelihood of ER versus ER+ breast cancers (OR = 3.32, 95 % CI 1.33–8.28), but were not significant for TNBC versus LumA cancers (Figs. 2, 3). High levels of IFNγ were associated with two to threefold increased odds of ER and TNBCs. Among postmenopausal women, high levels of pro-inflammatory IL-1β were associated with threefold reduced likelihood of both ER cancers (OR = 0.35, 95 % CI 0.17–0.73) and TNBC (OR = 0.32, 95 % CI 0.14–0.73), compared to ER+ or LumA. Postmenopausal women with high IL-10 levels, had reduced odds of ER breast cancer (OR = 0.42, 95 % CI = 0.20–0.86) and TNBC (OR = 0.45, 95 % CI 0.20–1.00).

Several cytokine ratios indicating a high Th1 cytokine to IL-5 ratio (i.e., IL-12-p70/IL-5, IL-12-p40/IL-5, TNFα/IL-5, IFNγ/IL-5, IFNα2/IL-5, CXCL10/IL-5) were inversely associated with both ER and TNBCs (Figs. 2, 3). Compared to women in the lowest tertile, women in the highest tertiles were approximately 2–2.5 times less likely to have ER breast cancer compared to ER+ cancers, and 2.5–3 times less likely to have TNBC compared to LumA cancers. Associations were strongest among premenopausal women, with women in the highest tertile approximately three times less likely to have ER disease and up to five times less likely to have TNBC (CXCL10/IL-5, OR = 0.18, 95 % CI, 0.07–0.49). Among postmenopausal women, high Th1cytokine/IL5 ratios were associated with approximately twofold reduced likelihood of ER disease and 2.5-fold reduced odds of TNBC compared to women with low ratios. High CXCL10/CCL7 ratios, an indicator of Th1/Th2 chemokine balance, were also inversely associated with ER and TNBC compared to ER+ and LumA cancers, respectively, but only among premenopausal women.

The ratio IFNα2/IL-4, also an indicator of Th1/Th2 balance, showed low correlations with IFNα2/IL-5 (spearmean r = 0.15, see Online Resource Table 4) and IFN γ/IL-5 (spearman r = 0.02), but was moderately correlated with IFNα2/IL-10 (spearman r = 0.43), IFNα2/IL-6 (spearman r = 0.42), and IFNα2/CCL7 (r = 0.44). Women in the highest tertile of IFNα2/IL-4, IFNα2/IL-6, IFNα2/IL-10, or IFNα2/CCL7 had a 1.5- to 2.5-fold greater likelihood of having ER versus ER+ breast cancer compared to women in the lowest tertile, with relationships being statistically significant only in premenopausal women, although tests of heterogeneity by menopausal status were only borderline statistically significant for IFNα2/IL-6 (p-int = 0.07). Ratios of IFNα2/IL-4, IFNα2/IL-10, IFNα2/IL-6, and IFNα2/TNFα, IFNα2/CCL7, and IFNα2/CCL11 were all positively associated with triple negative disease compared to LumA cancers, and high IL-1β/IL-1RA was associated with reduced likelihood, but only among postmenopausal women.

Discussion

This is the first study to examine relationships between circulating cytokines and breast cancer subtypes in a relatively large population. Our most significant finding was that high IL-5 levels, a Th2 cytokine, were associated with increased risk of ER compared to ER+ cancers and TNBCs compared to LumA cancer, particularly among premenopausal women. Pronounced Th1 immune activation, coupled with low levels of IL-5, was associated with the lowest risk of ER and TNBCs, compared to ER+ and LumA cancers, respectively, particularly among premenopausal women. High ratios of IFNα2 with other Th2-related cytokines (IL-4, IL-10, IL-6, CCL2, CCL7, CCL11), and TNFα, however, were associated with increased odds of ER and TNBCs compared to ER+ and LumA cancers. These findings, taken together, support the hypothesis that immune function is associated with development of ER and TNBC compared to ER+ and LumA cancers, and that immune pathways may be physiologically most relevant among younger women who are most likely to be diagnosed with these aggressive phenotypes.

Research on the possible role of cytokines in cancer has largely focused on the tumor microenvironment and their expression in tumor-associated macrophages, which are thought to play an important role in breast cancer progression [2730]. Few studies, however, have examined circulating cytokine levels and breast cancer risk, which are likely to yield insights into the role of underlying constitutional host immunity on carcinogenesis, and complement findings associated with tumor-driven immune changes. A limitation of previous studies [3135] has been small sample sizes, all involving fewer than 100 patients. Only one study, with only 53 women, examined relationships between circulating Th1 and Th2-related cytokines and breast cancer subtypes [36].

Of the 27 cytokines measured, high levels of IL-5 were most strongly associated with greater odds of ER and TNBC compared to ER+ and LumA cancers, respectively. IL-5 is produced by CD4+ Th2 lymphocytes and is specific in driving eosinophil-dependent inflammatory diseases typically associated with allergic diseases such as asthma and with parasitic worm (helminth) infections [3741]. GM-CSF, CCL3, CCL7, CXCL8, but not CCL11, as other cytokines involved in eosinophil chemotaxis [42] were also related to increased odds of ER and TNBC among premenopausal women, but not as strongly as for IL-5 (Online Resource Tables 2 and 3). Associations were strongest among premenopausal women, possibly because estrogens stimulate IL-5 production and eosinophil mobilization [43]. Exacerbation of asthma, for instance, is common during pregnancy [44]. A meta-analysis of atopic diseases with breast cancer risk, however, did not show any associations with asthma or “any allergy”, although effect estimates were not adjusted for effective management of these conditions as potential confounders that might attenuate risk estimates, and specific breast cancer subtypes were not considered [45].

Although our study was conducted in a Caucasian population, associations between immune factors, particularly IL5, and ER breast cancer could inform a better understanding of the high prevalence of ER breast cancer in African-American (AA) women, who also experience more allergies and asthma [1, 6, 4648]. The shift towards Th2 immunity among AAs is thought to be an evolutionary response to endemic exposure to helminths in sub-Saharan Africa [49, 50]. Previous observations that risk of TNBCs is associated with increasing parity may also, in part, be mediated by shifts towards Th2 immunity [6, 5153] since pregnancy is characterized by lower Th1 cytokines, increased Th2 cytokines, and a reduction in the Th1/Th2 ratio [5459]. Restoration to normal Th1/Th2 immunity occurs postpartum [60]. Interestingly, one study found decreased serum Th1/Th2 ratio, consistent with continued depressed cellular immunity, among formula-feeding mothers, but not among women who breastfed, suggesting that lactation may confer some immunological benefit to mothers [61]. This shift, possibly mediated by prolactin leads to Th1 activation [62, 63], may explain why lactation ameliorates the increased risk of ER and TNBC among African-Americans [6].

In our data, both ER and TNBCs were associated with higher levels of Th1 cytokines IFNγ, TNFα, and IFNα2, compared to ER+ and LumA cancers. Pro-inflammatory cytokines produced by CD4+ T cells enhance killer CD8+ T cells, have direct toxic effects on tumor cells, and can activate anti-angiogenic mechanisms [17, 64, 65]. These findings indicate a greater degree of immune activation, likely reflecting the increased disease aggressiveness of ER and TNBCs. Consistent with this interpretation, others have found IL-12 levels, the main cytokine regulating Th1 differentiation, to be higher in breast cancer patients compared to healthy controls, and correlated with more advanced disease stage [66, 67]. Impaired IFNα signaling has been observed in stage II or higher breast cancers and is hypothesized to be a common immune defect in human cancers [68]. As an early response cytokine, IFNα might be pivotal in cancer control because of its role in priming the host immune response, increasing natural killer (NK) cell cytotoxicity, and transitioning the immune system from an innate to an adaptive immune response through a number of mechanisms including increases in Th1/Th2 balance [6973].

The balance of Th1 and Th2 cell populations is implicated in a number of diseases, including cancer [7480]. A strong Th1 response is critical for effective antitumor immunity, whereas Th2 responses mediated by IL-4 can potentiate M2-bioactivity of tumor associated macrophages, leading to growth promotion and metastasis [17, 81]. In our study, high ratios of Th1 cytokines to the Th2 cytokine IL-5 (IL12/IL-5, IFNα2/IL-5, TNFα/IL-5, IFNγ/IL-5, CXCL10/IL-5) were strongly associated with reduced risk of ER compared to ER+ cancers or TNBCs compared to LumA cancers, but ratios of IFNα2 to other Th2 cytokines (IL-4, IL-6, IL-10, CCL2, CCL11) went in the opposite direction. The reasons for this are unclear, but may indicate more nuanced effects of Th2-related immunity on risk of ER and TNBC, including potential influences by other modulators of immune function such as macrophages, regulatory T cells and Th17 cells [8185]. IL-4, for instance, has pleiotropic effects and can inhibit basal and estrogen-induced cell proliferation and induce apoptosis in breast cancer cells [8689], and in certain situations promote a Th1 immune response [9093].

Our findings between circulating cytokines and ER and TNBCs are unlikely due to tumor-driven changes in plasma cytokine levels. Previous study on expression of cytokine levels in breast tumors suggest that Th1 and Th2 cytokines, including IFNγ and IL-5, are not major cytokines produced by tumors [9496], with IL-5 being non-detectable in both breast tumors and normal breast tissue [94]. Rather, several inflammatory cytokines produced by monocytes (IL-1β, IL-6, CCL2) or macrophages (such as IL-1β, IL-6, CXCL8, CCL4, and TNFα) appear to be more highly expressed in tumors [94, 9799], with inflammatory cytokines CXCL8, IL-6, and CCL2 levels being more highly expressed in ER cancers [94, 97, 98, 100, 101]. Nevertheless, given that our study design is cross-sectional, we cannot rule out the possibility that breast tumors or different breast cancer subtypes can potentially influence systemic immunity differently. Thus, our findings will need to be confirmed prospectively in a future study.

Our multivariate models did not adjust for lifestyle factors or comorbidities that are potentially associated with cytokine levels, such as body mass index and use of anti-inflammatory medications, because circulating cytokines may be within the causal pathway of these factors with respect to breast cancer risk. Thus, their inclusion in analytic models may lead to attenuation of effect estimates. The approach of examining circulating cytokine levels as a biomarker of immune phenotype without adjustments for factors that can potentially impact their levels also allowed for examination of “effective” exposure to these cytokines after accounting for multivariate influences, whether measured or unmeasured. Because disease-free controls were not included in this study, our findings show immune differences between ER and TNBCs in relation to less aggressive ER+ and LumA breast cancers. Future studies are required to determine if circulating cytokines, as contributors to immune regulation, are related to increased risk of either ER and/or ER+ disease compared to healthy women without breast cancer. A notable strength in our study, was the use of blood samples from treatment naïve breast cancer patients, which allowed us to examine differences in host immune phenotype unconfounded by treatment effects.

In summary, our findings show immune pathways to be important in ER and TNBCs particularly among younger women. Future prospective research involving a larger study population with samples drawn prior to disease diagnosis is needed to validate findings and determine if circulating cytokine levels are related to differences in immune cell profiles at the tumor site, and whether these immune-related differences predict response to therapy and/or disease prognosis. Another important area of investigation will be to determine whether our findings hold for African-American women, who are more likely to be diagnosed with ER and TNBCs. If future research establishes a role for IL-5 in the etiology of ER breast cancer, promising adjunct treatment strategies for ER and TNBC might include IFNα2, which increase Th1 versus Th2 balance, and have been clinically employed as antineoplastic therapeutic drugs [69, 72, 73], as well as IL-5-targeted therapies, such as the monoclonal antibodies that have been developed to neutralize IL-5 or target IL-5 R for the treatment of eosinophilic diseases [102105]. Our findings also indicate the possibility that among women diagnosed with ER or TNBC, who also have an underlying Th-2 related disease, such as asthma, an effort to optimally manage these conditions might result in improvements in breast cancer outcomes.

Supplementary Material

01

Acknowledgments

Funding for this research was provided by the Breast Cancer Research Foundation (PI: Christine Ambrosone). Dr. Song Yao was the recipient of a fellowship from the Department of Defense Breast Cancer Research Program (W81XWH-08-1-0223). Funding bodies for this study did not play any role in the collection, analysis, interpretation of data, in the writing of the manuscript, and in the decision to submit the manuscript for publication. The Roswell Park Cancer Institute Databank and Biorepository, the Clinical Data Network and Flow & Image Cytometry Department are CCSG Shared Resources (NIH P30 CA016056-27).

Abbreviations

AJCC

American Joint Committee on Cancer

BRCA1

Breast cancer type 1 susceptibility protein

CCL2

Chemokine (C–C motif) ligand 2

CCL7

Chemokine (C–C motif) ligand 7

CCL11

Chemokine (C–C motif) ligand 11

CK

Cytokeratin

CXCL10

Chemokine (C–X–C motif) ligand 10

DBBR

Data Bank and Biorepository

EGFR+

Epidermal growth factor receptor positive

ER

Estrogen receptor

ER+

Estrogen receptor positive

ER

Estrogen receptor negative

G-CSF

Granulocyte-colony stimulating factor

GM-CSF

Granulocyte macrophage-colony stimulating factor

HER2

Human epidermal growth factor receptor 2 negative

IFN

Interferon

IHC

Immunohistochemistry

IL

Interleukin

IL1RA

Interleukin-1 receptor antagonist

IP-10

Interferon gamma-induced protein 10

IRB

Institutional Review Board

LumA

Luminal A

MCP

Monocyte chemotactic protein

MDC

Macrophage-derived chemokine

MIP-1

Macrophage inflammatory protein-1

QC

Quality control

RPCI

Roswell Park Cancer Institute

TAMS

Tumor-associated macrophages

Th1

T-helper type 1

Th2

T-helper type 2

Th17

T-helper type 17

TNBC

Triple negative breast cancer

TNF

Tumor necrosis factor

Footnotes

Findings from this manuscript were presented at the AACR Post-GWAS Horizons in Molecular Epidemiology Conference in Hollywood FL, Nov 11–14, 2012.

Electronic supplementary material The online version of this article (doi:10.1007/s10549-013-2549-3) contains supplementary material, which is available to authorized users.

Conflicts of interest The authors have no potential conflicts of interest to disclose.

Contributor Information

Chi-Chen Hong, Email: Chi-Chen.Hong@RoswellPark.org, Department of Cancer Prevention and Control, Roswell Park Cancer Institute, Buffalo, NY 14263, USA.

Song Yao, Department of Cancer Prevention and Control, Roswell Park Cancer Institute, Buffalo, NY 14263, USA.

Susan E. McCann, Department of Cancer Prevention and Control, Roswell Park Cancer Institute, Buffalo, NY 14263, USA

Ree Y. Dolnick, Department of Flow & Image Cytometry, Roswell Park Cancer Institute, Buffalo, NY, USA

Paul K. Wallace, Department of Flow & Image Cytometry, Roswell Park Cancer Institute, Buffalo, NY, USA

Zhihong Gong, Department of Cancer Prevention and Control, Roswell Park Cancer Institute, Buffalo, NY 14263, USA.

Lei Quan, Department of Cancer Prevention and Control, Roswell Park Cancer Institute, Buffalo, NY 14263, USA.

Kelvin P. Lee, Department of Immunology, Roswell Park Cancer Institute, Buffalo, NY, USA

Sharon S. Evans, Department of Immunology, Roswell Park Cancer Institute, Buffalo, NY, USA

Elizabeth A. Repasky, Department of Immunology, Roswell Park Cancer Institute, Buffalo, NY, USA

Stephen B. Edge, Department of Surgical Oncology, Roswell Park Cancer Institute, Buffalo, NY, USA

Christine B. Ambrosone, Department of Cancer Prevention and Control, Roswell Park Cancer Institute, Buffalo, NY 14263, USA

References

  • 1.Carey LA, Perou CM, Livasy CA, et al. Race, breast cancer subtypes, and survival in the Carolina Breast Cancer Study. JAMA. 2006;295:2492–2502. doi: 10.1001/jama.295.21.2492. [DOI] [PubMed] [Google Scholar]
  • 2.Blows FM, Driver KE, Schmidt MK, et al. Subtyping of breast cancer by immunohistochemistry to investigate a relationship between subtype and short and long term survival: a collaborative analysis of data for 10,159 cases from 12 studies. PLoS Med. 2010;7:e1000279. doi: 10.1371/journal.pmed.1000279. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Tischkowitz M, Brunet JS, Begin LR, et al. Use of immunohistochemical markers can refine prognosis in triple negative breast cancer. BMC Cancer. 2007;7:134. doi: 10.1186/1471-2407-7-134. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Nofech-Mozes S, Trudeau M, Kahn HK, et al. Patterns of recurrence in the basal and non-basal subtypes of triple-negative breast cancers. Breast Cancer Res Treat. 2009;118:131–137. doi: 10.1007/s10549-008-0295-8. [DOI] [PubMed] [Google Scholar]
  • 5.Carey L, Winer E, Viale G, et al. Triple-negative breast cancer: disease entity or title of convenience? Nat Rev Clin Oncol. 2010;7:683–692. doi: 10.1038/nrclinonc.2010.154. [DOI] [PubMed] [Google Scholar]
  • 6.Millikan RC, Newman B, Tse CK, et al. Epidemiology of basal-like breast cancer. Breast Cancer Res Treat. 2008;109:123–139. doi: 10.1007/s10549-007-9632-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Hines LM, Risendal B, Byers T, et al. Ethnic disparities in breast tumor phenotypic subtypes in Hispanic and non-Hispanic white women. J Womens Health (Larchmt) 2011;20:1543–1550. doi: 10.1089/jwh.2010.2558. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Davis AA, Kaklamani VG. Metabolic syndrome and triple-negative breast cancer: a new paradigm. Int J Breast Cancer. 2012;2012:809291. doi: 10.1155/2012/809291. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Kwan ML, Kushi LH, Weltzien E, et al. Epidemiology of breast cancer subtypes in two prospective cohort studies of breast cancer survivors. Breast Cancer Res. 2009;11:R31. doi: 10.1186/bcr2261. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Pierce BL, Ballard-Barbash R, Bernstein L, et al. Elevated biomarkers of inflammation are associated with reduced survival among breast cancer patients. J Clin Oncol. 2009;27:3437–3444. doi: 10.1200/JCO.2008.18.9068. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Benoy IH, Salgado R, Van Dam P, et al. Increased serum interleukin-8 in patients with early and metastatic breast cancer correlates with early dissemination and survival. Clin Cancer Res. 2004;10:7157–7162. doi: 10.1158/1078-0432.CCR-04-0812. [DOI] [PubMed] [Google Scholar]
  • 12.Teschendorff AE, Miremadi A, Pinder SE, et al. An immune response gene expression module identifies a good prognosis subtype in estrogen receptor negative breast cancer. Genome Biol. 2007;8:R157. doi: 10.1186/gb-2007-8-8-r157. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Rody A, Karn T, Liedtke C, et al. A clinically relevant gene signature in triple negative and basal-like breast cancer. Breast Cancer Res. 20110;13:R97. doi: 10.1186/bcr3035. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Bianchini G, Qi Y, Alvarez RH, et al. Molecular anatomy of breast cancer stroma and its prognostic value in estrogen receptor-positive and -negative cancers. J Clin Oncol. 2010;28:4316–4323. doi: 10.1200/JCO.2009.27.2419. [DOI] [PubMed] [Google Scholar]
  • 15.DeNardo DG, Coussens LM. Inflammation and breast cancer. Balancing immune response: crosstalk between adaptive and innate immune cells during breast cancer progression. Breast Cancer Res. 2007;9:212. doi: 10.1186/bcr1746. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Bindea G, Mlecnik B, Fridman WH, et al. Natural immunity to cancer in humans. Curr Opin Immunol. 2010;22:215–222. doi: 10.1016/j.coi.2010.02.006. [DOI] [PubMed] [Google Scholar]
  • 17.Pardoll D. Metastasis-promoting immunity: when T cells turn to the dark side. Cancer Cell. 2009;16:81–82. doi: 10.1016/j.ccr.2009.07.007. [DOI] [PubMed] [Google Scholar]
  • 18.Waugh DJ, Wilson C. The interleukin-8 pathway in cancer. Clin Cancer Res. 2008;14:6735–6741. doi: 10.1158/1078-0432.CCR-07-4843. [DOI] [PubMed] [Google Scholar]
  • 19.Gabay C. Interleukin-6 and chronic inflammation. Arthritis Res Ther. 2006;8(Suppl 2):S3. doi: 10.1186/ar1917. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Salgado R, Junius S, Benoy I, et al. Circulating interleukin-6 predicts survival in patients with metastatic breast cancer. Int J Cancer. 2003;103:642–646. doi: 10.1002/ijc.10833. [DOI] [PubMed] [Google Scholar]
  • 21.Elenkov IJ. Glucocorticoids and the Th1/Th2 balance. Ann N Y Acad Sci. 2004;1024:138–146. doi: 10.1196/annals.1321.010. [DOI] [PubMed] [Google Scholar]
  • 22.Kidd P. Th1/Th2 balance: the hypothesis, its limitations, and implications for health and disease. Altern Med Rev. 2003;8:223–246. [PubMed] [Google Scholar]
  • 23.Onoe K, Yanagawa Y, Minami K, et al. Th1 or Th2 balance regulated by interaction between dendritic cells and NKT cells. Immunol Res. 2007;38:319–332. doi: 10.1007/s12026-007-0011-5. [DOI] [PubMed] [Google Scholar]
  • 24.Ambrosone CB, Nesline MK, Davis W. Establishing a cancer center data bank and biorepository for multidisciplinary research. Cancer Epidemiol Biomarkers Prev. 2006;15:1575–1577. doi: 10.1158/1055-9965.EPI-06-0628. [DOI] [PubMed] [Google Scholar]
  • 25.Yao S, Sucheston LE, Millen AE, et al. Pretreatment serum concentrations of 25-hydroxyvitamin D and breast cancer prognostic characteristics: a case-control and a case-series study. PLoS ONE. 2011;6:e17251. doi: 10.1371/journal.pone.0017251. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Benjamini Y, Hochberg Y. Controlling the false discovery rate: a practical and powerful approach to multiple testing. J Roy Stat Soc B. 1995;57:289–300. [Google Scholar]
  • 27.Laoui D, Movahedi K, Van Overmeire E, et al. Tumor-associated macrophages in breast cancer: distinct subsets, distinct functions. Int J Dev Biol. 2011;55:861–867. doi: 10.1387/ijdb.113371dl. [DOI] [PubMed] [Google Scholar]
  • 28.Sheu BC, Chang WC, Cheng CY, et al. Cytokine regulation networks in the cancer microenvironment. Front Biosci. 2008;13:6255–6268. doi: 10.2741/3152. [DOI] [PubMed] [Google Scholar]
  • 29.Wilson J, Balkwill F. The role of cytokines in the epithelial cancer microenvironment. Semin Cancer Biol. 2002;12:113–120. doi: 10.1006/scbi.2001.0419. [DOI] [PubMed] [Google Scholar]
  • 30.Goldberg JE, Schwertfeger KL. Proinflammatory cytokines in breast cancer: mechanisms of action and potential targets for therapeutics. Curr Drug Targets. 2010;11:1133–1146. doi: 10.2174/138945010792006799. [DOI] [PubMed] [Google Scholar]
  • 31.Lyon DE, McCain NL, Walter J, et al. Cytokine comparisons between women with breast cancer and women with a negative breast biopsy. Nurs Res. 2008;57:51–58. doi: 10.1097/01.NNR.0000280655.58266.6c. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Saglam S, Suzme R, Gurdol F. Serum tumor necrosis factor-alpha and interleukin-2 concentrations in newly diagnosed ERBB2 (HER2/neu) positive breast cancer patients. Int J Biol Markers. 2009;24:142–146. doi: 10.1177/172460080902400303. [DOI] [PubMed] [Google Scholar]
  • 33.Gonzalez RM, Daly DS, Tan R, et al. Plasma biomarker profiles differ depending on breast cancer subtype but RANTES is consistently increased. Cancer Epidemiol Biomarkers Prev. 2011;20:1543–1551. doi: 10.1158/1055-9965.EPI-10-1248. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Narita D, Seclaman E, Ursoniu S, et al. Expression of CCL18 and interleukin-6 in the plasma of breast cancer patients as compared with benign tumor patients and healthy controls. Rom J Morphol Embryol. 2011;52:1261–1267. [PubMed] [Google Scholar]
  • 35.Pusztai L, Mendoza TR, Reuben JM, et al. Changes in plasma levels of inflammatory cytokines in response to paclitaxel chemotherapy. Cytokine. 2004;25:94–102. doi: 10.1016/j.cyto.2003.10.004. [DOI] [PubMed] [Google Scholar]
  • 36.Herrera AC, Panis C, Victorino VJ, Campos FC, Colado-Simão AN, Cecchini AL, Cecchini R. Molecular subtype is determinant on inflammatory status and immunological profile from invasive breast cancer patients. Cancer Immunol Immunother. 2012;61(11):2193–2201. doi: 10.1007/s00262-012-1283-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Corren J. Inhibition of interleukin-5 for the treatment of eosinophilic diseases. Discov Med. 2012;13:305–312. [PubMed] [Google Scholar]
  • 38.Corren J. Anti-interleukin-5 antibody therapy in asthma and allergies. Curr Opin Allergy Clin Immunol. 2011;11:565–570. doi: 10.1097/ACI.0b013e32834c3d30. [DOI] [PubMed] [Google Scholar]
  • 39.Lee JJ, Jacobsen EA, McGarry MP, et al. Eosinophils in health and disease: the LIAR hypothesis. Clin Exp Allergy. 2010;40:563–575. doi: 10.1111/j.1365-2222.2010.03484.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Clutterbuck EJ, Hirst EM, Sanderson CJ. Human interleukin-5 (IL-5) regulates the production of eosinophils in human bone marrow cultures: comparison and interaction with IL-1, IL-3, IL-6, and GMCSF. Blood. 1989;73:1504–1512. [PubMed] [Google Scholar]
  • 41.Till S, Dickason R, Huston D, et al. IL-5 secretion by allergen-stimulated CD4+ T cells in primary culture: relationship to expression of allergic disease. J Allergy Clin Immunol. 1997;99:563–569. doi: 10.1016/s0091-6749(97)70085-x. [DOI] [PubMed] [Google Scholar]
  • 42.Simson L, Foster PS. Chemokine and cytokine cooperativity: eosinophil migration in the asthmatic response. Immunol Cell Biol. 2000;78:415–422. doi: 10.1046/j.1440-1711.2000.00922.x. [DOI] [PubMed] [Google Scholar]
  • 43.Cai Y, Zhou J, Webb DC. Estrogen stimulates Th2 cytokine production and regulates the compartmentalisation of eosinophils during allergen challenge in a mouse model of asthma. Int Arch Allergy Immunol. 2012;158:252–260. doi: 10.1159/000331437. [DOI] [PubMed] [Google Scholar]
  • 44.Katz O, Sheiner E. Asthma and pregnancy: a review of two decades. Expert Rev Respir Med. 2008;2:97–107. doi: 10.1586/17476348.2.1.97. [DOI] [PubMed] [Google Scholar]
  • 45.Vojtechova P, Martin RM. The association of atopic diseases with breast, prostate, and colorectal cancers: a metaanalysis. Cancer Causes Control. 2009;20:1091–1105. doi: 10.1007/s10552-009-9334-y. [DOI] [PubMed] [Google Scholar]
  • 46.Moorman JE, Rudd RA, Johnson CA, et al. National surveillance for asthma—United States, 1980–2004. MMWR Surveill Summ. 2007;56:1–54. [PubMed] [Google Scholar]
  • 47.Holgate ST. Innate and adaptive immune responses in asthma. Nat Med. 2012;18:673–683. doi: 10.1038/nm.2731. [DOI] [PubMed] [Google Scholar]
  • 48.Oliphant CJ, Barlow JL, McKenzie AN. Insights into the initiation of type 2 immune responses. Immunology. 2011;134:378–385. doi: 10.1111/j.1365-2567.2011.03499.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Anthony RM, Rutitzky LI, Urban JF, Jr, et al. Protective immune mechanisms in helminth infection. Nat Rev Immunol. 2007;7:975–987. doi: 10.1038/nri2199. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Rook GA. Review series on helminths, immune modulation and the hygiene hypothesis: the broader implications of the hygiene hypothesis. Immunology. 2009;126:3–11. doi: 10.1111/j.1365-2567.2008.03007.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Lyons TR, Schedin PJ, Borges VF. Pregnancy and breast cancer: when they collide. J Mammary Gland Biol Neoplasia. 2009;14:87–98. doi: 10.1007/s10911-009-9119-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.O’Brien J, Lyons T, Monks J, et al. Alternatively activated macrophages and collagen remodeling characterize the postpartum involuting mammary gland across species. Am J Pathol. 2010;176:1241–1255. doi: 10.2353/ajpath.2010.090735. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.O’Brien J, Schedin P. Macrophages in breast cancer: do involution macrophages account for the poor prognosis of pregnancy-associated breast cancer? J Mammary Gland Biol Neoplasia. 2009;14:145–157. doi: 10.1007/s10911-009-9118-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Reinhard G, Noll A, Schlebusch H, et al. Shifts in the TH1/TH2 balance during human pregnancy correlate with apoptotic changes. Biochem Biophys Res Commun. 1998;245:933–938. doi: 10.1006/bbrc.1998.8549. [DOI] [PubMed] [Google Scholar]
  • 55.Saito S, Tsukaguchi N, Hasegawa T, et al. Distribution of Th1, Th2, and Th0 and the Th1/Th2 cell ratios in human peripheral and endometrial T cells. Am J Reprod Immunol. 1999;42:240–245. doi: 10.1111/j.1600-0897.1999.tb00097.x. [DOI] [PubMed] [Google Scholar]
  • 56.Saito S, Sakai M, Sasaki Y, et al. Quantitative analysis of peripheral blood Th0, Th1, Th2 and the Th1:Th2 cell ratio during normal human pregnancy and preeclampsia. Clin Exp Immunol. 1999;117:550–555. doi: 10.1046/j.1365-2249.1999.00997.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Sykes L, MacIntyre DA, Yap XJ, et al. Changes in the Th1:Th2 cytokine bias in pregnancy and the effects of the anti-inflammatory cyclopentenone prostaglandin 15-deoxy-Delta(12,14)-prostaglandin J2. Mediators Inflamm. 2012;2012:416739. doi: 10.1155/2012/416739. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Luppi P. How immune mechanisms are affected by pregnancy. Vaccine. 2003;21:3352–3357. doi: 10.1016/s0264-410x(03)00331-1. [DOI] [PubMed] [Google Scholar]
  • 59.Rastogi D, Wang C, Lendor C, et al. T-helper type 2 polarization among asthmatics during and following pregnancy. Clin Exp Allergy. 2006;36:892–898. doi: 10.1111/j.1365-2222.2006.02519.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Halonen M, Lohman IC, Stern DA, et al. Th1/Th2 patterns and balance in cytokine production in the parents and infants of a large birth cohort. J Immunol. 2009;182:3285–3293. doi: 10.4049/jimmunol.0711996. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Groer MW, Davis MW. Cytokines, infections, stress, and dysphoric moods in breastfeeders and formula feeders. J Obstet Gynecol Neonatal Nurs. 2006;35:599–607. doi: 10.1111/j.1552-6909.2006.00083.x. [DOI] [PubMed] [Google Scholar]
  • 62.Dimitrov S, Lange T, Fehm HL, et al. A regulatory role of prolactin, growth hormone, and corticosteroids for human T-cell production of cytokines. Brain Behav Immun. 2004;18:368–374. doi: 10.1016/j.bbi.2003.09.014. [DOI] [PubMed] [Google Scholar]
  • 63.Meli R, Bentivoglio C, Nuzzo I, et al. Th1-Th2 response in hyperprolactinemic mice infected with Salmonella enterica serovar Typhimurium . Eur Cytokine Netw. 2003;14:186–191. [PubMed] [Google Scholar]
  • 64.Weiss JM, Subleski JJ, Wigginton JM, et al. Immunotherapy of cancer by IL-12-based cytokine combinations. Expert Opin Biol Ther. 2007;7:1705–1721. doi: 10.1517/14712598.7.11.1705. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Duda DG, Sunamura M, Lozonschi L, et al. Direct in vitro evidence and in vivo analysis of the antiangiogenesis effects of interleukin 12. Cancer Res. 2000;60:1111–1116. [PubMed] [Google Scholar]
  • 66.Kozlowski L, Zakrzewska I, Tokajuk P, et al. Concentration of interleukin-6 (IL-6), interleukin-8 (IL-8) and interleukin-10 (IL-10) in blood serum of breast cancer patients. Rocz Akad Med Bialymst. 2003;48:82–84. [PubMed] [Google Scholar]
  • 67.Hussein MZ, Al Fikky A, Abdel Bar I, et al. Serum IL-6 and IL-12 levels in breast cancer patients. Egypt J Immunol. 2004;11:165–170. [PubMed] [Google Scholar]
  • 68.Critchley-Thorne RJ, Simons DL, Yan N, et al. Impaired interferon signaling is a common immune defect in human cancer. Proc Natl Acad Sci USA. 2009;106:9010–9015. doi: 10.1073/pnas.0901329106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Brassard DL, Grace MJ, Bordens RW. Interferon-alpha as an immunotherapeutic protein. J Leukoc Biol. 2002;71:565–581. [PubMed] [Google Scholar]
  • 70.Biron CA. Role of early cytokines, including alpha and beta interferons (IFN-alpha/beta), in innate and adaptive immune responses to viral infections. Semin Immunol. 1998;10:383–390. doi: 10.1006/smim.1998.0138. [DOI] [PubMed] [Google Scholar]
  • 71.Wenner CA, Guler ML, Macatonia SE, et al. Roles of IFN-gamma and IFN-alpha in IL-12-induced T helper cell-1 development. J Immunol. 1996;156:1442–1447. [PubMed] [Google Scholar]
  • 72.Eguchi J, Hiroishi K, Ishii S, et al. Interferon-alpha and interleukin-12 gene therapy of cancer: interferon-alpha induces tumor-specific immune responses while interleukin-12 stimulates non-specific killing. Cancer Immunol Immunother. 2003;52:378–386. doi: 10.1007/s00262-002-0367-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Moschos SJ, Edington HD, Land SR, et al. Neoadjuvant treatment of regional stage IIIB melanoma with high-dose interferon alfa-2b induces objective tumor regression in association with modulation of tumor infiltrating host cellular immune responses. J Clin Oncol. 2006;24:3164–3171. doi: 10.1200/JCO.2005.05.2498. [DOI] [PubMed] [Google Scholar]
  • 74.Bleotu C, Chifiriuc MC, Grigore R, et al. Investigation of Th1/Th2 cytokine profiles in patients with laryngo-pharyngeal. HPV-positive cancers. Eur Arch Otorhinolaryngol. 2012 doi: 10.1007/s00405-012-2067-7. [DOI] [PubMed] [Google Scholar]
  • 75.Ubukata H, Motohashi G, Tabuchi T, et al. Evaluations of interferon-gamma/interleukin-4 ratio and neutrophil/lymphocyte ratio as prognostic indicators in gastric cancer patients. J Surg Oncol. 2010;102:742–747. doi: 10.1002/jso.21725. [DOI] [PubMed] [Google Scholar]
  • 76.Crane IJ, Forrester JV. Th1 and Th2 lymphocytes in autoimmune disease. Crit Rev Immunol. 2005;25:75–102. doi: 10.1615/critrevimmunol.v25.i2.10. [DOI] [PubMed] [Google Scholar]
  • 77.Ghoreschi K, Thomas P, Breit S, et al. Interleukin-4 therapy of psoriasis induces Th2 responses and improves human autoimmune disease. Nat Med. 2003;9:40–46. doi: 10.1038/nm804. [DOI] [PubMed] [Google Scholar]
  • 78.Nguyen TH, Casale TB. Immune modulation for treatment of allergic disease. Immunol Rev. 2011;242:258–271. doi: 10.1111/j.1600-065X.2011.01034.x. [DOI] [PubMed] [Google Scholar]
  • 79.Hamilton DH, Bretscher PA. Different immune correlates associated with tumor progression and regression: implications for prevention and treatment of cancer. Cancer Immunol Immunother. 2008;57:1125–1136. doi: 10.1007/s00262-007-0442-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Kennedy R, Celis E. Multiple roles for CD4+ T cells in anti-tumor immune responses. Immunol Rev. 2008;222:129–144. doi: 10.1111/j.1600-065X.2008.00616.x. [DOI] [PubMed] [Google Scholar]
  • 81.DeNardo DG, Barreto JB, Andreu P, et al. CD4(+) T cells regulate pulmonary metastasis of mammary carcinomas by enhancing protumor properties of macrophages. Cancer Cell. 2009;16:91–102. doi: 10.1016/j.ccr.2009.06.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Goodman WA, Levine AD, Massari JV, et al. IL-6 signaling in psoriasis prevents immune suppression by regulatory T cells. J Immunol. 2009;183:3170–3176. doi: 10.4049/jimmunol.0803721. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Oh SH, Park CO, Wu WH, et al. Corticotropin-releasing hormone downregulates IL-10 production by adaptive forkhead box protein 3-negative regulatory T cells in patients with atopic dermatitis. J Allergy Clin Immunol. 2012;129:151–159. e1–e6. doi: 10.1016/j.jaci.2011.09.008. [DOI] [PubMed] [Google Scholar]
  • 84.Chaudhry A, Samstein RM, Treuting P, et al. Interleukin-10 signaling in regulatory T cells is required for suppression of Th17 cell-mediated inflammation. Immunity. 2011;34:566–578. doi: 10.1016/j.immuni.2011.03.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Pace L, Pioli C, Doria G. IL-4 modulation of CD4+ CD25+ T regulatory cell-mediated suppression. J Immunol. 2005;174:7645–7653. doi: 10.4049/jimmunol.174.12.7645. [DOI] [PubMed] [Google Scholar]
  • 86.Gooch JL, Lee AV, Yee D. Interleukin 4 inhibits growth and induces apoptosis in human breast cancer cells. Cancer Res. 1998;58:4199–4205. [PubMed] [Google Scholar]
  • 87.Toi M, Bicknell R, Harris AL. Inhibition of colon and breast carcinoma cell growth by interleukin-4. Cancer Res. 1992;52:275–279. [PubMed] [Google Scholar]
  • 88.Gooch JL, Christy B, Yee D. STAT6 mediates interleu-kin-4 growth inhibition in human breast cancer cells. Neoplasia. 2002;4:324–331. doi: 10.1038/sj.neo.7900248. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Nagai S, Toi M. Interleukin-4 and breast cancer. Breast Cancer. 2000;7:181–186. doi: 10.1007/BF02967457. [DOI] [PubMed] [Google Scholar]
  • 90.Hochrein H, O’Keeffe M, Luft T, et al. Interleukin (IL)-4 is a major regulatory cytokine governing bioactive IL-12 production by mouse and human dendritic cells. J Exp Med. 2000;192:823–833. doi: 10.1084/jem.192.6.823. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Ebner S, Ratzinger G, Krosbacher B, et al. Production of IL-12 by human monocyte-derived dendritic cells is optimal when the stimulus is given at the onset of maturation, and is further enhanced by IL-4. J Immunol. 2001;166:633–641. doi: 10.4049/jimmunol.166.1.633. [DOI] [PubMed] [Google Scholar]
  • 92.Bream JH, Curiel RE, Yu CR, et al. IL-4 synergistically enhances both IL-2- and IL-12-induced IFN-gamma expression in murine NK cells. Blood. 2003;102:207–214. doi: 10.1182/blood-2002-08-2602. [DOI] [PubMed] [Google Scholar]
  • 93.Eguchi J, Hiroishi K, Ishii S, et al. Interleukin-4 gene transduced tumor cells promote a potent tumor-specific Th1-type response in cooperation with interferon-alpha transduction. Gene Ther. 2005;12:733–741. doi: 10.1038/sj.gt.3302401. [DOI] [PubMed] [Google Scholar]
  • 94.Chavey C, Bibeau F, Gourgou-Bourgade S, et al. Oestrogen receptor negative breast cancers exhibit high cytokine content. Breast Cancer Res. 2007;9:R15. doi: 10.1186/bcr1648. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Green AR, Green VL, White MC, et al. Expression of cytokine messenger RNA in normal and neoplastic human breast tissue: identification of interleukin-8 as a potential regulatory factor in breast tumours. Int J Cancer. 1997;72:937–941. doi: 10.1002/(sici)1097-0215(19970917)72:6<937::aid-ijc3>3.0.co;2-q. [DOI] [PubMed] [Google Scholar]
  • 96.Venetsanakos E, Beckman I, Bradley J, et al. High incidence of interleukin 10 mRNA but not interleukin 2 mRNA detected in human breast tumours. Br J Cancer. 1997;75:1826–1830. doi: 10.1038/bjc.1997.311. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Freund A, Chauveau C, Brouillet JP, et al. IL-8 expression and its possible relationship with estrogen-receptor-negative status of breast cancer cells. Oncogene. 2003;22:256–265. doi: 10.1038/sj.onc.1206113. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Lin Y, Huang R, Chen L, et al. Identification of interleukin-8 as estrogen receptor-regulated factor involved in breast cancer invasion and angiogenesis by protein arrays. Int J Cancer. 2004;109:507–515. doi: 10.1002/ijc.11724. [DOI] [PubMed] [Google Scholar]
  • 99.Soria G, Ofri-Shahak M, Haas I, et al. Inflammatory mediators in breast cancer: coordinated expression of TNFalpha & IL-1beta with CCL2 & CCL5 and effects on epithelial-to-mesenchymal transition. BMC Cancer. 2011;11:130. doi: 10.1186/1471-2407-11-130. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Pantschenko AG, Pushkar I, Anderson KH, et al. The interleukin-1 family of cytokines and receptors in human breast cancer: implications for tumor progression. Int J Oncol. 2003;23:269–284. [PubMed] [Google Scholar]
  • 101.Singer CF, Hudelist G, Gschwantler-Kaulich D, et al. Interleukin-1alpha protein secretion in breast cancer is associated with poor differentiation and estrogen receptor alpha negativity. Int J Gynecol Cancer. 2006;16(Suppl 2):556–559. doi: 10.1111/j.1525-1438.2006.00695.x. [DOI] [PubMed] [Google Scholar]
  • 102.Bochner BS, Gleich GJ. What targeting eosinophils has taught us about their role in diseases. J Allergy Clin Immunol. 2010;126:16–25. doi: 10.1016/j.jaci.2010.02.026. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Molfino NA, Gossage D, Kolbeck R, et al. Molecular and clinical rationale for therapeutic targeting of interleukin-5 and its receptor. Clin Exp Allergy. 2012;42:712–737. doi: 10.1111/j.1365-2222.2011.03854.x. [DOI] [PubMed] [Google Scholar]
  • 104.Kolbeck R, Kozhich A, Koike M, et al. MEDI-563, a humanized anti-IL-5 receptor alpha mAb with enhanced antibody-dependent cell-mediated cytotoxicity function. J Allergy Clin Immunol. 2010;125(1344–1353):e2. doi: 10.1016/j.jaci.2010.04.004. [DOI] [PubMed] [Google Scholar]
  • 105.Busse WW, Katial R, Gossage D, et al. Safety profile, pharmacokinetics, and biologic activity of MEDI-563, an anti-IL-5 receptor alpha antibody, in a phase I study of subjects with mild asthma. J Allergy Clin Immunol. 2010;125(1237–1244):e2. doi: 10.1016/j.jaci.2010.04.005. [DOI] [PubMed] [Google Scholar]
  • 106.Zamarron BF, Chen W. Dual roles of immune cells and their factors in cancer development and progression. Int J Biol Sci. 2011;7:651–658. doi: 10.7150/ijbs.7.651. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Biswas SK, Mantovani A. Macrophage plasticity and interaction with lymphocyte subsets: cancer as a paradigm. Nat Immunol. 2010;11:889–896. doi: 10.1038/ni.1937. [DOI] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

01

RESOURCES