Skip to main content
Erschienen in: Journal of Translational Medicine 1/2019

Open Access 01.12.2019 | Research

Clinical verification of the relationship between smoking and the immune microenvironment of breast cancer

verfasst von: Koji Takada, Shinichiro Kashiwagi, Yuka Asano, Wataru Goto, Katsuyuki Takahashi, Hisakazu Fujita, Tsutomu Takashima, Shuhei Tomita, Kosei Hirakawa, Masaichi Ohira

Erschienen in: Journal of Translational Medicine | Ausgabe 1/2019

Abstract

Background

The immune tumor microenvironment (iTME) is thought to affect the response to chemotherapy, and tumor-infiltrating lymphocytes (TILs) are often used as an indicator to evaluate the iTME. Smoking is involved in carcinogenesis, the relationship between smoking and the iTME of lung cancer has been reported. We hypothesized that smoking would affect the iTME of breast cancer and aimed to examine this relationship based on the amount of pre-diagnosis smoking and the subsequent effects on treatment response and prognosis.

Methods

This retrospective study evaluated data from 149 patients who underwent preoperative chemotherapy for triple-negative or HER2-enriched breast cancer. TILs were assessed in biopsy specimens at diagnosis. The data of all patients were used to calculate each patient’s smoking amount based on pack-years.

Results

Relative to the low smoking group, the high smoking group had a significant greater TILs density (p = 0.043) and a significantly better pathological complete response (pCR) rate (p = 0.042). However, there was no significant difference according to smoking amount in disease-free survival (p = 0.114) or overall survival (p = 0.347).

Conclusions

Smoking may influence the iTME, with an activated iTME being associated with pCR rate. Therefore, controlled activation of the microenvironment in this setting may help improve patients’ prognosis.
Hinweise

Electronic supplementary material

The online version of this article (https://​doi.​org/​10.​1186/​s12967-019-1773-y) contains supplementary material, which is available to authorized users.
Abkürzungen
iTME
immune tumor microenvironment
TILs
tumor-infiltrating lymphocytes
pCR
pathological complete response
DFS
disease-free survival
OS
overall survival
TNBC
triple-negative breast cancer
HER2BC
human epidermal growth factor receptor 2-enriched breast cancer
POC
preoperative chemotherapy
ER
estrogen receptor
PgR
progesterone receptor
HER2
human epidermal growth factor receptor 2
ORR
objective response rate
pCR
pathological complete response
HR
hazard ratio
CI
confidence interval
REMARK
Reporting Recommendations for Tumor Marker Prognostic Studies
FOXP3
forkhead box protein 3

Background

The immune tumor microenvironment (iTME) is thought to affect the response to chemotherapy, and tumor-infiltrating lymphocytes (TILs) are often used as an indicator to evaluate the iTME [13]. Many studies have revealed that a high TILs density in breast cancer is associated with good therapeutic effects, such as pathological complete response (pCR), prolonged disease-free survival (DFS), and prolonged overall survival (OS) [4, 5]. It became commonly known that affect TILs density in breast cancer is the cancer subtype, with many reports indicating that a high TILs density is associated with high-risk subtypes, such as triple-negative breast cancer (TNBC) and human epidermal growth factor receptor 2-enriched breast cancer (HER2BC) [6, 7]. In recent years, it has also been reported that special genes affect TILs, and it is also important to examine the relationship between genes and the iTME [8, 9].
Smoking is involved in the genesis of many carcinomas, including breast cancer [8], with the carcinogenic substances in tobacco smoke causing chronic inflammatory conditions in the microvessels [10, 11]. Recent studies have also indicated that the iTME is deeply involved in carcinogenesis and that chronic inflammation promotes this process [12, 13]. The relationship between smoking and the iTME of lung cancer has been reported [5, 14], although no reports have examined the relationship between smoking and the iTME of breast cancer. Therefore, we hypothesized that smoking would affect the iTME of breast cancer and aimed to examine this relationship based on the amount of pre-diagnosis smoking and the subsequent effects on treatment response and prognosis.

Methods

Patient background

This retrospective study evaluated data from 149 patients who underwent preoperative chemotherapy (POC) for resectable TNBC or HER2BC between February 2007 and December 2017 at the Osaka City University Hospital. All patients were questioned regarding their smoking history at the initial visit (cigarettes smoked per day and years of smoking), and the data were used to calculate each patient’s smoking amount based on pack-years (Table 1). The breast cancers were diagnosed pathologically and classified according to subtype based on the immunohistochemical expression of estrogen receptor (ER), progesterone receptor (PgR), human epidermal growth factor receptor 2 (HER2), and K-i67. Cases were defined as either HER2BC (ER−, PgR−, and HER2+) or TNBC (ER−, PgR−, and HER2−).
Table 1
Pack-years of smoking
To calculate smoking pack-years:
  Divide the number of cigarettes smoked per day by 20 (the number of cigarettes in a pack)
  Then multiply by the number of years smoked
ex. 1
(70 cigarettes/day ÷ 20 cigarettes/pack) × 10 years = 35 pack-years
ex. 2
(35 cigarettes/day ÷ 20 cigarettes/pack) × 20 years = 35 pack-years
ex. 3
(20 cigarettes/day ÷ 20 cigarettes/pack) × 20 years = 20 pack-years
All patients received a standardized outpatient POC regimen that consisted of four courses of FEC100 (fluorouracil: 500 mg/m2, epirubicin: 100 mg/m2, and cyclophosphamide: 500 mg/m2) every 3 weeks, which was followed by 12 courses of weekly paclitaxel (80 mg/m2). The patients with HER2BC also received trastuzumab during the paclitaxel treatment as a weekly dose (2 mg/kg) or tri-weekly dose (6 mg/kg) [1517]. Staging and therapeutic effect were evaluated using ultrasonography, computed tomography, and bone scintigraphy based on the Response Evaluation Criteria in Solid Tumors [18]. Patients who achieved clinically partial or complete response were categorized as “responders” in the objective response rate (ORR), while patients with clinically stable or progressive disease were defined as “non-responders”. The patients subsequently underwent mastectomy or breast-conserving surgery [19], and the pathological therapeutic effect of the POC was evaluated using the resected specimens. Pathological complete response (pCR) was defined as complete disappearance of the lesion’s invasive components, including the lymph nodes, with or without intraductal components, according to the National Surgical Adjuvant Breast and Bowel Project B-18 protocol [20]. All patients received postoperative radiotherapy delivered to the remnant breast, and the standard postoperative adjuvant therapy was selected based on the cancer subtype. Patients were followed-up after surgery to detect recurrence using physical examinations every 3 months, ultrasonography every 6 months, and computed tomography and bone scintigraphy annually. The DFS interval was calculated from the day of surgery to the first instance of recurrence or death, while OS was calculated from the day of surgery to death.

Histopathological evaluation of TILs density

Specimens that were used to pathologically diagnose breast cancer (obtained via core needle biopsy or vacuum-assisted biopsy) were used to determine the TILs density. In the present study, TILs were defined as lymphocytes infiltrating within the tumor stroma [21]. The TILs density was calculated as the average from five randomly selected fields, and the results were classified as a score of 3 (> 50%), a score of 2 (11–50%), a score of 1 (≤ 10%), or a score of 0 (absent) (Additional file 1: Fig. S1). Based on previous reports [22, 23], we defined a high TILs density as scores of 2–3 (i.e., > 10%) and a low TILs density as scores of 0–1 (≤ 10%).

Statistical analysis

All analyses were performed using JMP software (version 11; SAS Institute, Cary, NC). Differences in the study variables were evaluated using the Chi square test or Fisher’s exact test, as appropriate. The Kaplan–Meier method was used to estimate the DFS and OS outcomes, which were compared using the log-rank test. A Cox proportional hazards model was used to calculate hazard ratios (HRs) and 95% confidence intervals (CIs), and multivariable analysis was performed using a Cox regression model and the backward stepwise selection method. Differences were considered statistically significant at p-values of < 0.05.

Ethics statement

This study was conducted at the Osaka City University Graduate School of Medicine (Osaka, Japan) according to the Reporting Recommendations for Tumor Marker Prognostic Studies (REMARK) guidelines. The study protocol involved a retrospectively written research, pathological evaluation, and statistical analysis plan [24]. The study complied with the provisions of the Declaration of Helsinki, and all patients provided written informed consent for their treatment and data collection. The study’s retrospective protocol was approved by the ethics committee of Osaka City University (#926).

Results

Clinicopathological features

The clinicopathological features of the 149 women are listed in Table 2. The median age at surgery was 56 years (range 24–75 years old). The median follow-up duration was 1288 days after surgery (range 13–3615 days). The median tumor diameter was 27.6 mm (range 10.2–98.0 mm) and 98 patients (65.8%) were diagnosed with N1–3 lymph node metastasis based on their imaging results. Sixty-two patients had HER2BC (41.6%) and 87 patients had TNBC (58.4%). Ninety-one patients (61.1%) had a high TILs density and 58 patients (38.9%) had a low TILs density at their diagnosis. One hundred and five patients (70.5%) reported never smoking, and 44 patients (29.5%) reported a median smoking amount of 20 pack-years (range 2.5–135 pack-years). Based on the receiver operating characteristic curve analysis, the optimal smoking cut-off value for predicting DFS was defined as 2.5 pack-years, which yielded a distribution of 43 patients (28.9%) in the high smoking group and 106 patients (71.1%) in the low-smoking group (area under the curve: 0.588, sensitivity: 0.325, specificity: 0.846) (Additional file 2: Fig. S2). The ORR was 82.6% and 74 patients (49.7%) achieved a pCR. The therapeutic response was significantly higher among patients with HER2BC than among patients with TNBC (p = 0.023) (Table 3). However, there were no significant differences in the two groups’ pCR rates (p = 0.085), TILs density (p = 0.206), or smoking amount (p = 0.134).
Table 2
Clinicopathological features of 149 patients who were treated with preoperative chemotherapy
Parameters (n = 149)
Number of patients (%)
Age (years old)
56 (24–75)
Tumour size (mm)
27.6 (10.2–98.0)
Skin infiltration
 Negative/positive
134 (89.9%)/15 (10.1%)
Lymph node metastasis
 N0/N1/N2/N3
51 (34.2%)/54 (36.2%)/29 (19.5%)/15 (10.1%)
HER2
 Negative/positive
87 (58.4%)/62 (41.6%)
Ki67
 Negative/positive
23 (15.4%)/126 (84.6%)
ORR
 Non-responders/responders
11 (7.4%)/138 (92.6%)
pCR
 Negative/positive
75 (50.3%)/74 (49.7%)
Recurrence
 Negative/positive
123 (82.6%)/26 (17.4%)
TILs
 Low/high
58 (38.9%)/91 (61.1%)
Smoker
 No/yes
105 (70.5%)/44 (29.5%)
Pack-years of smokers
20 (2.5–135)
Pack-years
 Low/high
106 (71.1%)/43 (28.9%)
HER human epidermal growth factor receptor, ORR objective response rate, pCR pathological complete response, TILs tumour-infiltrating lymphocytes
Table 3
Comparison of clinicopathological features by subtype
Parameters
Intrinsic subtype
p value
HER2-enriched breast cancer (n = 62)
Triple-negative breast cancer (n = 87)
Age (years old)
 ≤ 56
26 (41.9%)
49 (56.3%)
0.085
 > 56
36 (58.1%)
38 (43.7%)
Tumour size (mm)
 ≤ 27.6
30 (48.4%)
45 (51.7%)
0.690
 > 27.6
32 (51.6%)
42 (48.3%)
Skin infiltration
 Negative
54 (87.1%)
80 (92.0%)
0.335
 Positive
8 (12.9%)
7 (8.0%)
Lymph node status
 Negative
25 (40.3%)
26 (29.9%)
0.188
 Positive
37 (59.7%)
61 (70.1%)
Ki67
 Negative
14 (22.6%)
9 (10.3%)
0.042
 Positive
48 (77.4%)
78 (89.7%)
ORR
 Non-responders
1 (1.6%)
10 (11.5%)
0.023
 Responders
61 (98.4%)
77 (88.5%)
pCR
 Negative
26 (41.9%)
49 (56.3%)
0.085
 Positive
36 (58.1%)
38 (43.7%)
Recurrence
 Negative
55 (88.7%)
68 (78.2%)
0.096
 Positive
7 (11.3%)
19 (21.8%)
TILs
 Low
20 (32.3%)
38 (43.7%)
0.206
 High
42 (67.7%)
49 (56.3%)
Pack-years
 Low
40 (64.5%)
66 (75.9%)
0.134
 High
22 (35.5%)
21 (24.1%)
HER human epidermal growth factor receptor, ORR objective response rate, pCR pathological complete response, TILs tumour-infiltrating lymphocytes

The associations of smoking with clinicopathological features, DFS, and OS

Table 4 shows the results of the associations between smoking and the patients’ clinicopathological features. No significant correlation was found between comparing smokers and never smokers. However, when divided into two groups according to smoking amount, correlation with clinicopathological features was recognized. Relative to the low smoking group, the high smoking group had a significant greater TILs density (p = 0.043) and a significantly better pCR rate (p = 0.042). In the univariate analysis, prolonged DFS was significantly associated with pCR (p < 0.001, HR 0.203, 95% CI 0.068–0.499) and a high TILs density (p = 0.001, HR 0.252, 95% CI 0.107–0.553) (Table 5). In addition, prolonged OS was significantly associated with pCR (p = 0.002, HR 0.183, 95% CI 0.042–0.561) and a high TILs density (p = 0.035, HR 0.357, 95% CI 00.129–0.929) (Table 5). However, there was no significant difference according to smoking amount in DFS (p = 0.114) or OS (p = 0.347) (Fig. 1).
Table 4
Difference in clinicopathological features due to pack-years
Parameters
Smoker
p value
Pack-years
p value
Yes (n = 44)
No (n = 105)
High (n = 43)
Low (n = 106)
Age (years old)
 ≤ 56
24 (54.5%)
51 (48.6%)
0.509
24 (55.8%)
51 (48.1%)
0.398
 > 56
20 (45.5%)
54 (51.4%)
19 (44.2%)
55 (51.9%)
Tumour size (mm)
 ≤ 27.6
25 (56.8%)
50 (47.6%)
0.309
25 (58.1%)
50 (47.2%)
0.228
 > 27.6
19 (43.2%)
55 (52.4%)
18 (41.9%)
56 (52.8%)
Skin infiltration
 Negative
40 (90.9%)
94 (89.5%)
0.799
40 (93.0%)
94 (88.7%)
0.428
 Positive
4 (9.1%)
11 (10.5%)
3 (7.0%)
12 (11.3%)
Lymph node status
 Negative
18 (40.9%)
33 (31.4%)
0.269
18 (41.9%)
33 (31.1%)
0.214
 Positive
26 (59.1%)
72 (68.6%)
25 (58.1%)
73 (68.9%)
Ki67
 Negative
6 (13.6%)
17 (16.2%)
0.69
6 (14.0%)
17 (16.0%)
0.752
 Positive
38 (86.4%)
88 (83.8%)
 
37 (86.0%)
89 (84.0%)
 
Intrinsic subtype
 HER2-enriched
22 (50.0%)
40 (38.1%)
0.181
22 (51.2%)
40 (37.7%)
0.134
 Triple-negative
22 (50.0%)
65 (61.9%)
21 (48.8%)
66 (62.3%)
ORR
 Non-responders
3 (6.8%)
8 (7.6%)
0.866
3 (7.0%)
8 (7.5%)
0.905
 Responders
41 (93.2%)
97 (92.4%)
40 (93.0%)
98 (92.5%)
pCR
 Negative
17 (38.6%)
58 (55.2%)
0.065
16 (37.2%)
59 (55.7%)
0.042
 Positive
27 (61.4%)
47 (44.8%)
 
27 (62.8%)
47 (44.3%)
 
Recurrence
 Negative
40 (90.9%)
83 (79.0%)
0.083
39 (90.7%)
84 (79.2%)
0.096
 Positive
4 (9.1%)
22 (21.0%)
4 (9.3%)
22 (20.8%)
TILs
 Low
13 (29.6%)
45 (42.9%)
0.075
12 (27.9%)
46 (43.4%)
0.043
 High
31 (70.5%)
60 (57.1%)
31 (72.1%)
60 (56.6%)
HER human epidermal growth factor receptor, ORR objective response rate, pCR pathological complete response, TILs tumour-infiltrating lymphocytes
Table 5
Univariate and multivariate analysis with respect to disease-free survival and overall survival
Parameters
Univariate analysis
Multivariate analysis
Hazard ratio
95% CI
p value
Hazard ratio
95% CI
p value
Disease-free survival
 Age at operation (year)
  ≤ 56
0.614
0.269–1.335
0.220
   
  > 56
 Tumour size (mm)
  ≤ 27.6
1.137
0.525–2.503
0.744
   
  > 27.6
 Skin infiltration
  Negative
1.556
0.455–4.067
0.440
   
  Positive
 Lymph node status
  Negative
2.440
0.933–8.343
0.071
1.677
0.617–5.859
0.331
  Positive
 Ki67
  Negative
0.394
0.180–0.926
0.034
0.770
0.321–1.942
0.568
  Positive
 Intrinsic subtype
  HER2-enriched
1.884
0.828–4.823
0.135
   
  Triple-negative
 ORR
  Non-responders
0.083
0.035–0.210
< 0.001
0.154
0.059–0.426
0.001
  Responders
 Pathological response
  Non-pCR
0.203
0.068–0.499
< 0.001
0.381
0.118–1.059
0.065
  pCR
 TILs
  Low
0.252
0.107–0.553
0.001
0.424
0.167–1.032
0.059
  High
 Pack-years
  Low
0.434
0.127–1.134
0.092
0.567
0.160–1.555
0.289
  High
Overall survival
 Age at operation (year)
  ≤ 56
0.508
0.175–1.338
0.172
   
  > 56
 Tumour size (mm)
  ≤ 27.6
1.123
0.429–2.993
0.811
   
  > 27.6
 Skin infiltration
  Negative
1.939
0.446–5.977
0.335
   
  Positive
 Lymph node status
  Negative
2.778
0.781–17.657
0.125
   
  Positive
      
 Ki67
  Negative
0.638
0.234–2.023
0.419
   
  Positive
 Intrinsic subtype
  HER2-enriched
1.610
0.597–5.060
0.357
   
  Triple-negative
 ORR
  Non-responders
0.077
0.026–0.238
< 0.001
0.451
0.044–0.520
0.004
  Responders
 Pathological response
  Non-pCR
0.183
0.042–0.561
0.002
0.282
0.062–0.953
0.041
  pCR
 TILs
  Low
0.357
0.129–0.929
0.035
0.634
0.212–1.861
0.403
  High
 Pack-years
  Low
0.554
0.128–1.700
0.325
   
  High
OS overall survival, CI confidence intervals, HER human epidermal growth factor receptor, ORR objective response rate, pCR pathological complete response, TILs tumour-infiltrating lymphocytes

Discussion

Smoking is a risk factor for various carcinomas, including breast cancer [25]. Smoking-related carcinogenesis is linked to various factors, with some of the components in tobacco smoke having estrogenic effects and others having antiestrogenic effects [26, 27]. Moreover, tobacco components can be carried through the blood to the mammary gland tissues where they cause DNA damage [10, 11]. Some researchers have indicated that smoking is associated with the development of ER+ breast cancer, while many others have reported that smoking is associated with ER− breast cancer [2631]. These differences may be related to race [27], which would be consistent with our findings, as all of our patients were Japanese and had ER− cancers. Furthermore, tissue culture and animal experiments have indicated that tobacco smoke components increase proliferative capacity and cause malignant transformation [3234], which further highlights the relationship between smoking and the development of TNBC or HER2BC.
The present study indicated that the HER2BC and TNBC subtypes were related to smoking and the cancer’s pre-treatment iTME. Interestingly, previous reports have indicated that a high TILs density was significantly associated with prolonged DFS and OS [4, 5], and the present study indicated that TILs density was associated with the pre-diagnosis smoking amount. These results indicate that local microimmune reactions are activated by chronic inflammation in microvessels, which may be related to the release of antigens as a result of smoking-related DNA damage. Given that a higher smoking amount was associated with a high TILs density, it is possible that smoking was related to the high pCR rate.
Although no previous studies have evaluated the relationship between smoking and the iTME in breast cancer, that relationship has been studied in lung cancer. For example, in non-small cell lung cancer, smoking was not associated with the expression of CD3, CD4, forkhead box protein 3 (FOXP3), and CD20, although smoking was associated with increased CD8 expression [14, 35]. Furthermore, increased numbers of CD8+ T-cells is associated with a good prognosis among patients with non-small cell lung cancer [14, 36]. Moreover, CD8 is a marker for cytotoxic T-cells, which are associated with an improved prognosis among patients with breast cancer [2, 37]. Although the present study did not directly evaluate the correlation between smoking and DFS or OS, the overall exposure to tobacco smoke is known to be associated with the risks of breast cancer recurrence, breast cancer-related death, and overall mortality [38, 39]. In this context, smoking could activate the iTME and affect the short-term therapeutic effect (i.e., pCR rate), although it might not be associated with the long-term therapeutic effect (i.e., DFS or OS) because it is not correlated with low oxygen levels caused by microangiopathy or deterioration of the iTME.
The present study has several limitations. First, the smoking amount was retrospective determined using self-reported data from at the patient’s diagnosis. Second, we did not consider smoking status after diagnosis or second-hand smoke, although passive smoking is an important risk factor for carcinogenesis [25] and lifelong exposure to smoke is more strongly related to the risks of carcinogenesis and recurrence (vs. current smoking status) [38, 39]. It is also reported that special genes, such as MAPKs/TP53, are affecting the iTME [8, 9]. That is, the iTME is also strongly related to genes. Since this result has only been investigated retrospectively, it is necessary to further examine the relationship between smoking and iTME with such as immunohistochemical staining, gene analysis or experiments in vitro. Moreover, it will be important to consider complete smoking-related data to examine the association of smoking with long-term prognosis among patients with breast cancer.

Conclusions

In conclusion, smoking may influence the iTME, with an activated iTME being associated with pCR rate. Therefore, controlled activation of the microenvironment in this setting may help improve patients’ prognosis.

Authors’ contributions

All authors were involved in the preparation of this manuscript. KTakada collected the data, and wrote the manuscript. SK, YA, WG, KTakahashi and TT performed the operation and designed the study. KTakada, SK and ST summarized the data and revised the manuscript. HF, KH and MO substantial contribution to the study design, performed the operation, and revised the manuscript. All authors read and approved the final manuscript.

Acknowledgements

We thank Yayoi Matsukiyo and Tomomi Okawa (Department of Surgical Oncology, Osaka City University Graduate School of Medicine) for helpful advice regarding data management.

Competing interests

The authors declare that they have no competing interests.

Availability of data and materials

The datasets supporting the conclusions of this article is included within the article.
Not applicable.
Written informed consent was obtained from all subjects. This research conformed to the provisions of the Declaration of Helsinki in 2013. All patients were informed of the investigational nature of this study and provided their written, informed consent. The study protocol was approved by the Ethics Committee of Osaka City University (#926).

Funding

This study was supported in part by Grants-in Aid for Scientific Research (KAKENHI, Nos. 25461992, 26461957, and 17K10559) from the Ministry of Education, Science, Sports, Culture and Technology of Japan.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://​creativecommons.​org/​licenses/​by/​4.​0/​), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated.
Literatur
1.
Zurück zum Zitat Zitvogel L, Kepp O, Kroemer G. Immune parameters affecting the efficacy of chemotherapeutic regimens. Nat Rev Clin Oncol. 2011;8(3):151–60.CrossRef Zitvogel L, Kepp O, Kroemer G. Immune parameters affecting the efficacy of chemotherapeutic regimens. Nat Rev Clin Oncol. 2011;8(3):151–60.CrossRef
2.
Zurück zum Zitat Fridman WH, Pages F, Sautes-Fridman C, Galon J. The immune contexture in human tumors: impact on clinical outcome. Nat Rev Cancer. 2012;12(4):298–306.CrossRef Fridman WH, Pages F, Sautes-Fridman C, Galon J. The immune contexture in human tumors: impact on clinical outcome. Nat Rev Cancer. 2012;12(4):298–306.CrossRef
3.
Zurück zum Zitat Couzin-Frankel J. Breakthrough of the year 2013. Cancer immunotherapy. Science. 2013;342(6165):1432–3.CrossRef Couzin-Frankel J. Breakthrough of the year 2013. Cancer immunotherapy. Science. 2013;342(6165):1432–3.CrossRef
4.
Zurück zum Zitat Adams S, Gray RJ, Demaria S, Goldstein L, Perez EA, Shulman LN, Martino S, Wang M, Jones VE, Saphner TJ, et al. Prognostic value of tumor-infiltrating lymphocytes in triple-negative breast cancers from two phase III randomized adjuvant breast cancer trials: ECOG 2197 and ECOG 1199. J Clin Oncol. 2014;32(27):2959–66.CrossRef Adams S, Gray RJ, Demaria S, Goldstein L, Perez EA, Shulman LN, Martino S, Wang M, Jones VE, Saphner TJ, et al. Prognostic value of tumor-infiltrating lymphocytes in triple-negative breast cancers from two phase III randomized adjuvant breast cancer trials: ECOG 2197 and ECOG 1199. J Clin Oncol. 2014;32(27):2959–66.CrossRef
5.
Zurück zum Zitat Denkert C, von Minckwitz G, Brase JC, Sinn BV, Gade S, Kronenwett R, Pfitzner BM, Salat C, Loi S, Schmitt WD, et al. Tumor-infiltrating lymphocytes and response to neoadjuvant chemotherapy with or without carboplatin in human epidermal growth factor receptor 2-positive and triple-negative primary breast cancers. J Clin Oncol. 2015;33(9):983–91.CrossRef Denkert C, von Minckwitz G, Brase JC, Sinn BV, Gade S, Kronenwett R, Pfitzner BM, Salat C, Loi S, Schmitt WD, et al. Tumor-infiltrating lymphocytes and response to neoadjuvant chemotherapy with or without carboplatin in human epidermal growth factor receptor 2-positive and triple-negative primary breast cancers. J Clin Oncol. 2015;33(9):983–91.CrossRef
6.
Zurück zum Zitat Ohtani H, Mori-Shiraishi K, Nakajima M, Ueki H. Defining lymphocyte-predominant breast cancer by the proportion of lymphocyte-rich stroma and its significance in routine histopathological diagnosis. Pathol Int. 2015;65(12):644–51.CrossRef Ohtani H, Mori-Shiraishi K, Nakajima M, Ueki H. Defining lymphocyte-predominant breast cancer by the proportion of lymphocyte-rich stroma and its significance in routine histopathological diagnosis. Pathol Int. 2015;65(12):644–51.CrossRef
7.
Zurück zum Zitat Stanton SE, Adams S, Disis ML. Variation in the incidence and magnitude of tumor-infiltrating lymphocytes in breast cancer subtypes: a systematic review. JAMA Oncol. 2016;2(10):1354–60.CrossRef Stanton SE, Adams S, Disis ML. Variation in the incidence and magnitude of tumor-infiltrating lymphocytes in breast cancer subtypes: a systematic review. JAMA Oncol. 2016;2(10):1354–60.CrossRef
8.
Zurück zum Zitat Loi S, Dushyanthen S, Beavis PA, Salgado R, Denkert C, Savas P, Combs S, Rimm DL, Giltnane JM, Estrada MV, et al. RAS/MAPK activation is associated with reduced tumor-infiltrating lymphocytes in triple-negative breast cancer: therapeutic cooperation between MEK and PD-1/PD-L1 immune checkpoint inhibitors. Clin Cancer Res. 2016;22(6):1499–509.CrossRef Loi S, Dushyanthen S, Beavis PA, Salgado R, Denkert C, Savas P, Combs S, Rimm DL, Giltnane JM, Estrada MV, et al. RAS/MAPK activation is associated with reduced tumor-infiltrating lymphocytes in triple-negative breast cancer: therapeutic cooperation between MEK and PD-1/PD-L1 immune checkpoint inhibitors. Clin Cancer Res. 2016;22(6):1499–509.CrossRef
9.
Zurück zum Zitat Quigley D, Silwal-Pandit L, Dannenfelser R, Langerød A, Vollan HK, Vaske C, Siegel JU, Troyanskaya O, Chin SF, Caldas C, et al. Lymphocyte invasion in IC10/basal-like breast tumors is associated with wild-type TP53. Mol Cancer Res. 2015;13(3):493–501.CrossRef Quigley D, Silwal-Pandit L, Dannenfelser R, Langerød A, Vollan HK, Vaske C, Siegel JU, Troyanskaya O, Chin SF, Caldas C, et al. Lymphocyte invasion in IC10/basal-like breast tumors is associated with wild-type TP53. Mol Cancer Res. 2015;13(3):493–501.CrossRef
10.
Zurück zum Zitat Ambrosone CB, Kropp S, Yang J, Yao S, Shields PG, Chang-Claude J. Cigarette smoking, N-acetyltransferase 2 genotypes, and breast cancer risk: pooled analysis and meta-analysis. Cancer Epidemiol Biomarkers Prev. 2008;17(1):15–26.CrossRef Ambrosone CB, Kropp S, Yang J, Yao S, Shields PG, Chang-Claude J. Cigarette smoking, N-acetyltransferase 2 genotypes, and breast cancer risk: pooled analysis and meta-analysis. Cancer Epidemiol Biomarkers Prev. 2008;17(1):15–26.CrossRef
11.
Zurück zum Zitat Hecht SS. Tobacco smoke carcinogens and breast cancer. Environ Mol Mutagen. 2002;39(2–3):119–26.CrossRef Hecht SS. Tobacco smoke carcinogens and breast cancer. Environ Mol Mutagen. 2002;39(2–3):119–26.CrossRef
12.
Zurück zum Zitat Hanahan D, Weinberg RA. Hallmarks of cancer: the next generation. Cell. 2011;144(5):646–74.CrossRef Hanahan D, Weinberg RA. Hallmarks of cancer: the next generation. Cell. 2011;144(5):646–74.CrossRef
13.
Zurück zum Zitat Hanahan D, Coussens LM. Accessories to the crime: functions of cells recruited to the tumor microenvironment. Cancer Cell. 2012;21(3):309–22.CrossRef Hanahan D, Coussens LM. Accessories to the crime: functions of cells recruited to the tumor microenvironment. Cancer Cell. 2012;21(3):309–22.CrossRef
14.
Zurück zum Zitat Schalper KA, Brown J, Carvajal-Hausdorf D, McLaughlin J, Velcheti V, Syrigos KN, Herbst RS, Rimm DL. Objective measurement and clinical significance of TILs in non-small cell lung cancer. J Natl Cancer Inst. 2015;107(3):dju435.CrossRef Schalper KA, Brown J, Carvajal-Hausdorf D, McLaughlin J, Velcheti V, Syrigos KN, Herbst RS, Rimm DL. Objective measurement and clinical significance of TILs in non-small cell lung cancer. J Natl Cancer Inst. 2015;107(3):dju435.CrossRef
15.
Zurück zum Zitat Mauri D, Pavlidis N, Ioannidis JP. Neoadjuvant versus adjuvant systemic treatment in breast cancer: a meta-analysis. J Natl Cancer Inst. 2005;97(3):188–94.CrossRef Mauri D, Pavlidis N, Ioannidis JP. Neoadjuvant versus adjuvant systemic treatment in breast cancer: a meta-analysis. J Natl Cancer Inst. 2005;97(3):188–94.CrossRef
16.
Zurück zum Zitat Mieog JS, van der Hage JA, van de Velde CJ. Preoperative chemotherapy for women with operable breast cancer. Cochrane Database Syst Rev. 2007;2:CD005002. Mieog JS, van der Hage JA, van de Velde CJ. Preoperative chemotherapy for women with operable breast cancer. Cochrane Database Syst Rev. 2007;2:CD005002.
17.
Zurück zum Zitat Kawajiri H, Takashima T, Onoda N, Kashiwagi S, Noda S, Ishikawa T, Wakasa K, Hirakawa K. Efficacy and feasibility of neoadjuvant chemotherapy with FEC 100 followed by weekly paclitaxel for operable breast cancer. Oncol Lett. 2012;4(4):612–6.CrossRef Kawajiri H, Takashima T, Onoda N, Kashiwagi S, Noda S, Ishikawa T, Wakasa K, Hirakawa K. Efficacy and feasibility of neoadjuvant chemotherapy with FEC 100 followed by weekly paclitaxel for operable breast cancer. Oncol Lett. 2012;4(4):612–6.CrossRef
18.
Zurück zum Zitat Eisenhauer EA, Therasse P, Bogaerts J, Schwartz LH, Sargent D, Ford R, Dancey J, Arbuck S, Gwyther S, Mooney M, et al. New response evaluation criteria in solid tumors: revised RECIST guideline (version 1.1). Eur J Cancer. 2009;45(2):228–47.CrossRef Eisenhauer EA, Therasse P, Bogaerts J, Schwartz LH, Sargent D, Ford R, Dancey J, Arbuck S, Gwyther S, Mooney M, et al. New response evaluation criteria in solid tumors: revised RECIST guideline (version 1.1). Eur J Cancer. 2009;45(2):228–47.CrossRef
19.
Zurück zum Zitat Kashiwagi S, Onoda N, Asano Y, Kurata K, Morisaki T, Noda S, Kawajiri H, Takashima T, Hirakawa K. Partial mastectomy using manual blunt dissection (MBD) in early breast cancer. BMC Surg. 2015;15:117.CrossRef Kashiwagi S, Onoda N, Asano Y, Kurata K, Morisaki T, Noda S, Kawajiri H, Takashima T, Hirakawa K. Partial mastectomy using manual blunt dissection (MBD) in early breast cancer. BMC Surg. 2015;15:117.CrossRef
20.
Zurück zum Zitat Wolmark N, Wang J, Mamounas E, Bryant J, Fisher B. Preoperative chemotherapy in patients with operable breast cancer: nine-year results from National Surgical Adjuvant Breast and Bowel Project B-18. J Natl Cancer Inst Monogr. 2001;30:96–102.CrossRef Wolmark N, Wang J, Mamounas E, Bryant J, Fisher B. Preoperative chemotherapy in patients with operable breast cancer: nine-year results from National Surgical Adjuvant Breast and Bowel Project B-18. J Natl Cancer Inst Monogr. 2001;30:96–102.CrossRef
21.
Zurück zum Zitat Salgado R, Denkert C, Demaria S, Sirtaine N, Klauschen F, Pruneri G, Wienert S, Van den Eynden G, Baehner FL, Penault-Llorca F, et al. The evaluation of tumor-infiltrating lymphocytes (TILs) in breast cancer: recommendations by an International TILs Working Group 2014. Ann Oncol. 2015;26(2):259–71.CrossRef Salgado R, Denkert C, Demaria S, Sirtaine N, Klauschen F, Pruneri G, Wienert S, Van den Eynden G, Baehner FL, Penault-Llorca F, et al. The evaluation of tumor-infiltrating lymphocytes (TILs) in breast cancer: recommendations by an International TILs Working Group 2014. Ann Oncol. 2015;26(2):259–71.CrossRef
22.
Zurück zum Zitat Kashiwagi S, Tsujio G, Asano Y, Goto W, Takada K, Takahashi K, Morisaki T, Fujita H, Takashima T, Tomita S, et al. Study on the progression types of cancer in patients with breast cancer undergoing eribulin chemotherapy and tumor microenvironment. J Transl Med. 2018;16(1):54.CrossRef Kashiwagi S, Tsujio G, Asano Y, Goto W, Takada K, Takahashi K, Morisaki T, Fujita H, Takashima T, Tomita S, et al. Study on the progression types of cancer in patients with breast cancer undergoing eribulin chemotherapy and tumor microenvironment. J Transl Med. 2018;16(1):54.CrossRef
23.
Zurück zum Zitat Kashiwagi S, Asano Y, Goto W, Takada K, Takahashi K, Noda S, Takashima T, Onoda N, Tomita S, Ohsawa M, et al. Use of tumor-infiltrating lymphocytes (TILs) to predict the treatment response to eribulin chemotherapy in breast cancer. PLoS ONE. 2017;12(2):e0170634.CrossRef Kashiwagi S, Asano Y, Goto W, Takada K, Takahashi K, Noda S, Takashima T, Onoda N, Tomita S, Ohsawa M, et al. Use of tumor-infiltrating lymphocytes (TILs) to predict the treatment response to eribulin chemotherapy in breast cancer. PLoS ONE. 2017;12(2):e0170634.CrossRef
24.
Zurück zum Zitat McShane LM, Altman DG, Sauerbrei W, Taube SE, Gion M, Clark GM. Statistics subcommittee of the NCIEWGoCD: reporting recommendations for tumor marker prognostic studies. J Clin Oncol. 2005;23(36):9067–72.CrossRef McShane LM, Altman DG, Sauerbrei W, Taube SE, Gion M, Clark GM. Statistics subcommittee of the NCIEWGoCD: reporting recommendations for tumor marker prognostic studies. J Clin Oncol. 2005;23(36):9067–72.CrossRef
25.
Zurück zum Zitat Dossus L, Boutron-Ruault MC, Kaaks R, Gram IT, Vilier A, Fervers B, Manjer J, Tjonneland A, Olsen A, Overvad K, et al. Active and passive cigarette smoking and breast cancer risk: results from the EPIC cohort. Int J Cancer. 2014;134(8):1871–88.CrossRef Dossus L, Boutron-Ruault MC, Kaaks R, Gram IT, Vilier A, Fervers B, Manjer J, Tjonneland A, Olsen A, Overvad K, et al. Active and passive cigarette smoking and breast cancer risk: results from the EPIC cohort. Int J Cancer. 2014;134(8):1871–88.CrossRef
26.
Zurück zum Zitat Kawai M, Malone KE, Tang MT, Li CI. Active smoking and the risk of estrogen receptor-positive and triple-negative breast cancer among women ages 20 to 44 years. Cancer. 2014;120(7):1026–34.CrossRef Kawai M, Malone KE, Tang MT, Li CI. Active smoking and the risk of estrogen receptor-positive and triple-negative breast cancer among women ages 20 to 44 years. Cancer. 2014;120(7):1026–34.CrossRef
27.
Zurück zum Zitat Park SY, Palmer JR, Rosenberg L, Haiman CA, Bandera EV, Bethea TN, Troester MA, Viscidi E, Kolonel LN, Olshan AF, et al. A case–control analysis of smoking and breast cancer in African American women: findings from the AMBER Consortium. Carcinogenesis. 2016;37(6):607–15.CrossRef Park SY, Palmer JR, Rosenberg L, Haiman CA, Bandera EV, Bethea TN, Troester MA, Viscidi E, Kolonel LN, Olshan AF, et al. A case–control analysis of smoking and breast cancer in African American women: findings from the AMBER Consortium. Carcinogenesis. 2016;37(6):607–15.CrossRef
28.
Zurück zum Zitat Morabia A, Bernstein M, Ruiz J, Heritier S, Diebold Berger S, Borisch B. Relation of smoking to breast cancer by estrogen receptor status. Int J Cancer. 1998;75(3):339–42.CrossRef Morabia A, Bernstein M, Ruiz J, Heritier S, Diebold Berger S, Borisch B. Relation of smoking to breast cancer by estrogen receptor status. Int J Cancer. 1998;75(3):339–42.CrossRef
29.
Zurück zum Zitat Manjer J, Malina J, Berglund G, Bondeson L, Garne JP, Janzon L. Smoking associated with hormone receptor negative breast cancer. Int J Cancer. 2001;91(4):580–4.CrossRef Manjer J, Malina J, Berglund G, Bondeson L, Garne JP, Janzon L. Smoking associated with hormone receptor negative breast cancer. Int J Cancer. 2001;91(4):580–4.CrossRef
30.
Zurück zum Zitat Nishino Y, Minami Y, Kawai M, Fukamachi K, Sato I, Ohuchi N, Kakugawa Y. Cigarette smoking and breast cancer risk in relation to joint estrogen and progesterone receptor status: a case–control study in Japan. Springerplus. 2014;3:65.CrossRef Nishino Y, Minami Y, Kawai M, Fukamachi K, Sato I, Ohuchi N, Kakugawa Y. Cigarette smoking and breast cancer risk in relation to joint estrogen and progesterone receptor status: a case–control study in Japan. Springerplus. 2014;3:65.CrossRef
31.
Zurück zum Zitat Cooper JA, Rohan TE, Cant EL, Horsfall DJ, Tilley WD. Risk factors for breast cancer by oestrogen receptor status: a population-based case–control study. Br J Cancer. 1989;59(1):119–25.CrossRef Cooper JA, Rohan TE, Cant EL, Horsfall DJ, Tilley WD. Risk factors for breast cancer by oestrogen receptor status: a population-based case–control study. Br J Cancer. 1989;59(1):119–25.CrossRef
32.
Zurück zum Zitat Nishioka T, Kim HS, Luo LY, Huang Y, Guo J, Chen CY. Sensitization of epithelial growth factor receptors by nicotine exposure to promote breast cancer cell growth. Breast Cancer Res. 2011;13(6):R113.CrossRef Nishioka T, Kim HS, Luo LY, Huang Y, Guo J, Chen CY. Sensitization of epithelial growth factor receptors by nicotine exposure to promote breast cancer cell growth. Breast Cancer Res. 2011;13(6):R113.CrossRef
33.
Zurück zum Zitat Melendez-Colon VJ, Luch A, Seidel A, Baird WM. Cancer initiation by polycyclic aromatic hydrocarbons results from formation of stable DNA adducts rather than apurinic sites. Carcinogenesis. 1999;20(10):1885–91.CrossRef Melendez-Colon VJ, Luch A, Seidel A, Baird WM. Cancer initiation by polycyclic aromatic hydrocarbons results from formation of stable DNA adducts rather than apurinic sites. Carcinogenesis. 1999;20(10):1885–91.CrossRef
34.
Zurück zum Zitat Forteza RM, Casalino-Matsuda SM, Falcon NS, Valencia Gattas M, Monzon ME. Hyaluronan and layilin mediate loss of airway epithelial barrier function induced by cigarette smoke by decreasing E-cadherin. J Biol Chem. 2012;287(50):42288–98.CrossRef Forteza RM, Casalino-Matsuda SM, Falcon NS, Valencia Gattas M, Monzon ME. Hyaluronan and layilin mediate loss of airway epithelial barrier function induced by cigarette smoke by decreasing E-cadherin. J Biol Chem. 2012;287(50):42288–98.CrossRef
35.
Zurück zum Zitat Kinoshita T, Muramatsu R, Fujita T, Nagumo H, Sakurai T, Noji S, Takahata E, Yaguchi T, Tsukamoto N, Kudo-Saito C, et al. Prognostic value of tumor-infiltrating lymphocytes differs depending on histological type and smoking habit in completely resected non-small-cell lung cancer. Ann Oncol. 2016;27(11):2117–23.CrossRef Kinoshita T, Muramatsu R, Fujita T, Nagumo H, Sakurai T, Noji S, Takahata E, Yaguchi T, Tsukamoto N, Kudo-Saito C, et al. Prognostic value of tumor-infiltrating lymphocytes differs depending on histological type and smoking habit in completely resected non-small-cell lung cancer. Ann Oncol. 2016;27(11):2117–23.CrossRef
36.
Zurück zum Zitat Donnem T, Hald SM, Paulsen EE, Richardsen E, Al-Saad S, Kilvaer TK, Brustugun OT, Helland A, Lund-Iversen M, Poehl M, et al. Stromal CD8 + T-cell density-A promising supplement to TNM staging in non-small cell lung cancer. Clin Cancer Res. 2015;21(11):2635–43.CrossRef Donnem T, Hald SM, Paulsen EE, Richardsen E, Al-Saad S, Kilvaer TK, Brustugun OT, Helland A, Lund-Iversen M, Poehl M, et al. Stromal CD8 + T-cell density-A promising supplement to TNM staging in non-small cell lung cancer. Clin Cancer Res. 2015;21(11):2635–43.CrossRef
37.
Zurück zum Zitat Asano Y, Kashiwagi S, Goto W, Kurata K, Noda S, Takashima T, Onoda N, Tanaka S, Ohsawa M, Hirakawa K. Tumor-infiltrating CD8 to FOXP3 lymphocyte ratio in predicting treatment responses to neoadjuvant chemotherapy of aggressive breast cancer. Br J Surg. 2016;103(7):845–54.CrossRef Asano Y, Kashiwagi S, Goto W, Kurata K, Noda S, Takashima T, Onoda N, Tanaka S, Ohsawa M, Hirakawa K. Tumor-infiltrating CD8 to FOXP3 lymphocyte ratio in predicting treatment responses to neoadjuvant chemotherapy of aggressive breast cancer. Br J Surg. 2016;103(7):845–54.CrossRef
38.
Zurück zum Zitat Saquib N, Stefanick ML, Natarajan L, Pierce JP. Mortality risk in former smokers with breast cancer: pack-years vs. smoking status. Int J Cancer. 2013;133(10):2493–7.CrossRef Saquib N, Stefanick ML, Natarajan L, Pierce JP. Mortality risk in former smokers with breast cancer: pack-years vs. smoking status. Int J Cancer. 2013;133(10):2493–7.CrossRef
39.
Zurück zum Zitat Pierce JP, Patterson RE, Senger CM, Flatt SW, Caan BJ, Natarajan L, Nechuta SJ, Poole EM, Shu XO, Chen WY. Lifetime cigarette smoking and breast cancer prognosis in the after breast cancer pooling project. J Natl Cancer Inst. 2014;106(1):djt359.CrossRef Pierce JP, Patterson RE, Senger CM, Flatt SW, Caan BJ, Natarajan L, Nechuta SJ, Poole EM, Shu XO, Chen WY. Lifetime cigarette smoking and breast cancer prognosis in the after breast cancer pooling project. J Natl Cancer Inst. 2014;106(1):djt359.CrossRef
Metadaten
Titel
Clinical verification of the relationship between smoking and the immune microenvironment of breast cancer
verfasst von
Koji Takada
Shinichiro Kashiwagi
Yuka Asano
Wataru Goto
Katsuyuki Takahashi
Hisakazu Fujita
Tsutomu Takashima
Shuhei Tomita
Kosei Hirakawa
Masaichi Ohira
Publikationsdatum
01.12.2019
Verlag
BioMed Central
Erschienen in
Journal of Translational Medicine / Ausgabe 1/2019
Elektronische ISSN: 1479-5876
DOI
https://doi.org/10.1186/s12967-019-1773-y

Weitere Artikel der Ausgabe 1/2019

Journal of Translational Medicine 1/2019 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Echinokokkose medikamentös behandeln oder operieren?

06.05.2024 DCK 2024 Kongressbericht

Die Therapie von Echinokokkosen sollte immer in spezialisierten Zentren erfolgen. Eine symptomlose Echinokokkose kann – egal ob von Hunde- oder Fuchsbandwurm ausgelöst – konservativ erfolgen. Wenn eine Op. nötig ist, kann es sinnvoll sein, vorher Zysten zu leeren und zu desinfizieren. 

Umsetzung der POMGAT-Leitlinie läuft

03.05.2024 DCK 2024 Kongressbericht

Seit November 2023 gibt es evidenzbasierte Empfehlungen zum perioperativen Management bei gastrointestinalen Tumoren (POMGAT) auf S3-Niveau. Vieles wird schon entsprechend der Empfehlungen durchgeführt. Wo es im Alltag noch hapert, zeigt eine Umfrage in einem Klinikverbund.

Proximale Humerusfraktur: Auch 100-Jährige operieren?

01.05.2024 DCK 2024 Kongressbericht

Mit dem demographischen Wandel versorgt auch die Chirurgie immer mehr betagte Menschen. Von Entwicklungen wie Fast-Track können auch ältere Menschen profitieren und bei proximaler Humerusfraktur können selbst manche 100-Jährige noch sicher operiert werden.

Die „Zehn Gebote“ des Endokarditis-Managements

30.04.2024 Endokarditis Leitlinie kompakt

Worauf kommt es beim Management von Personen mit infektiöser Endokarditis an? Eine Kardiologin und ein Kardiologe fassen die zehn wichtigsten Punkte der neuen ESC-Leitlinie zusammen.

Update Innere Medizin

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.