Skip to main content
Erschienen in: European Journal of Medical Research 1/2023

Open Access 01.12.2023 | Research

Clinical verification of the relationship between serum lipid metabolism and immune activity in breast cancer patients treated with neoadjuvant chemotherapy

verfasst von: Wataru Goto, Shinichiro Kashiwagi, Koji Takada, Yuka Asano, Kana Ogisawa, Tamami Morisaki, Masatsune Shibutani, Hiroaki Tanaka, Kiyoshi Maeda

Erschienen in: European Journal of Medical Research | Ausgabe 1/2023

Abstract

Background

Lipid metabolism has been recently reported to affect the prognosis and tumor immune activity in cancer patients. However, the effect of lipid metabolism on chemosensitivity in patients with breast cancer treated with neoadjuvant chemotherapy (NAC) remains unclear.

Methods

We examined 327 patients with breast cancer who were treated with NAC followed by curative surgery. The correlations between the serum levels of total cholesterol (TC) and triglyceride (TG) and the clinicopathological features, including the efficacy of NAC, neutrophil-to-lymphocyte ratio (NLR), and absolute lymphocyte count (ALC), were evaluated retrospectively.

Results

Serum TG levels were increased after NAC in all the subtypes, and the rate of change was the highest, especially in triple-negative breast cancer (TNBC) (21.0% → 48.1%). In addition, only TNBC patients with an objective response (OR) had significantly higher TG levels after NAC than those without (P = 0.049). Patients with a high ALC before NAC had significantly higher TG levels after NAC than patients with all breast cancer (P = 0.001), HER2-enriched breast cancer (P = 0.021), and TNBC (P = 0.008). Patients with a low NLR before NAC had significantly higher TG levels after NAC only among patients with TNBC (P = 0.025). In patients with human epidermal growth factor receptor 2-enriched breast cancer, the group with normal TC levels before NAC had significantly better OS than those with high TC levels (P = 0.013, log-rank test), and in patients with TNBC, the group with high TC levels after NAC had significantly better OS than those with normal TC levels (P = 0.014, log-rank test).

Conclusions

Good systemic immune activity and chemosensitivity may be associated with lipid metabolism regulated by NAC in TNBC patients.
Begleitmaterial
Hinweise

Supplementary Information

The online version contains supplementary material available at https://​doi.​org/​10.​1186/​s40001-022-00964-w.

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Abkürzungen
ALC
Absolute lymphocyte count
BMI
Body mass index
CR
Complete response
ER
Estrogen receptor
HER2
Human-epidermal growth factor receptor 2
NAC
Neoadjuvant chemotherapy
NLR
Neutrophil-to-lymphocyte ratio
OR
Objective response
ORR
Objective response rate
OS
Overall survival
pCR
Pathological complete response
PgR
Progesterone receptor
RFS
Recurrence-free survival
TC
Total-cholesterol
TG
Triglyceride
TNBC
Triple-negative breast cancer

Background

Neoadjuvant chemotherapy (NAC) is the gold standard treatment for breast cancer and its use has increased the rate of breast-conserving surgery [1, 2]. In addition, the pathological complete response (pCR) after NAC is a predictor of good outcome, and its prognostic value is greatest in aggressive subtypes, human epidermal growth factor receptor 2 (HER2)-enriched, and triple-negative breast cancer (TNBC) [3]. These intrinsic breast cancer subtypes have a high malignancy and immune activity. We have reported previously that immune-related biomarkers, including the neutrophil–lymphocyte ratio (NLR) or tumor-infiltrating lymphocytes in biopsy specimens before NAC, are associated significantly with high pCR rates in these breast cancer subtypes [4].
Immunometabolism has become a relatively new field in cancer immunotherapy, and it has been recognized that the regulation of metabolism can enhance antitumor immunity [57]. In the case of lipid metabolism, obesity has been shown to be associated positively with breast cancer risk in postmenopausal hormone receptor-positive women [8, 9]. In addition, while epidemiological evidence has shown no association between the use of agents for dyslipidemia, mainly statins, and reduced breast cancer incidence, it supports a protective effect of these drugs on reducing breast cancer recurrence or mortality [10]. Moreover, several basic studies have shown that statins can suppress cancer cell proliferation, exert anti-angiogenic effects, and reduce the invasiveness and metastatic potential of breast cancer cells [1116]. In addition, Crocetto et al. reported that the lipid alterations may be a potential tumor biomarker to detect bladder cancer, endocrine-related cancer, in clinical practice [17]; however, the relationship between lipid metabolism and immune activity in breast cancer patients has not been sufficiently investigated.
On the other hand, chemotherapy enhances antitumor immune responses [18, 19]. Some studies have revealed that changes in the lymphocytic subpopulations after NAC can be used as prognostic markers in patients with breast cancer patients [2023]. In contrast, other studies have examined the metabolic changes before and after chemotherapy in breast cancer patients and showed significant changes in lipid levels [24, 25]. Furthermore, Tian et al. reported that NAC exerts an adverse effect on lipid levels during chemotherapy [26].
The present study investigated the correlation between lipid metabolism, antitumor immune responses, and chemosensitivity in patients with breast cancer treated with NAC.

Methods

Patient background

Data from the Osaka City University Graduate School of Medicine (Osaka, Japan) between April 2007 and March 2018 were analyzed. A total of 351 patients were diagnosed with early stage breast cancer (stage IIA, IIB, IIIA) and underwent with primary systemic treatment and curative surgery. We excluded 24 patients treated with neoadjuvant endocrine therapy and included 327 patients treated with NAC in this study. T and N factors and tumor stage were stratified based on the TNM Classification, UICC Seventh Edition [27]. Tumors were classified into intrinsic subtypes according to the immunohistochemical expression of the estrogen receptor (ER), progesterone receptor (PgR), and HER2. We defined ER + and/or PgR + and HER2-breast cancer as luminal, ER + and/or PgR + and HER2 + breast cancer as luminal-HER2, ER- and PgR-, HER2 + breast cancer as HER2-enriched, and ER-, PgR-, and HER2-breast cancer as TNBC. The antitumor effect was assessed according to the Response Evaluation Criteria in Solid Tumors [28]. The objective response (OR) was calculated as the sum of the clinical partial response and complete response (CR). All the patients underwent mastectomy or breast-conserving surgery after NAC. The pCR was defined as the complete disappearance of the invasive compartment of the lesion with or without intraductal components, including the lymph nodes” [29]. Postoperative adjuvant therapy suitable for each intrinsic breast cancer subtype was performed, and standard postoperative radiotherapy to the remnant breast was administered if necessary. All patients underwent physical examinations, blood tests, ultrasonography, computed tomography, and bone scintigraphy scans. Overall survival (OS) was defined as the time from curative surgery to death from any cause, and recurrence-free survival (RFS) was defined as freedom from all locoregional and distant recurrences. The median follow-up time for the assessment of OS was 5.5 years (range 0.2–12.4 years) and for RFS was 4.9 years (range 0.1–12.0 years).

Blood sample analysis

Peripheral blood samples were obtained at the time of diagnosis, and preoperative blood samples were obtained within a week before surgery. We evaluated the serum lipid levels, including total cholesterol (TC) levels [categorized as low (≤ 149 mg/dl), normal (150–219 mg/dl), and high (≥ 220 mg/dl)] and triglyceride (TG) levels [categorized as low (≤ 49 mg/dl), normal (50–149 mg/dl), and high (≥ 150 mg/dl)]. The differential white blood cell counts were analyzed using a Coulter LH 750 Hematology Analyzer (Beckman Coulter, Brea, CA, USA). The neutrophil-to-lymphocyte ratio (NLR) was calculated from the blood samples by dividing the absolute neutrophil count by the absolute lymphocyte count (ALC).

Statistical analyses

Statistical analyses were performed using the JMP13 software (SAS Institute, Cary, NC, USA). Associations among the variables were analyzed using the χ2 or Fisher’s exact test, as appropriate. OS and RFS were estimated using the Kaplan–Meier method and log-rank test. Statistical significance was set at P < 0.05.

Results

Clinicopathological responses of all the breast cancer patients to NAC

The differences in clinicopathological features due to intrinsic breast cancer subtypes are presented in Table 1. A total of 327 patients were included in this study. Among these, 108 (33.0%), 42 (12.9%), 72 (22.0%), and 105 (32.1%) had luminal, luminal-HER2, HER2-enriched, and TNBC, respectively. NAC-related pCR was observed in 121 patients (37.0%). The evaluation based on the clinicopathological features revealed that the pCR rate was significantly higher in HER2-enriched (59.7%, 43/72) and TNBC patients (44.8%, 47/105) (P < 0.001). OR was observed in 295 patients (90.2%). The OR rate was high in all the breast cancer subtypes, and no significant differences were observed (P = 0.070). Patients with high TC levels increased after NAC in each breast cancer subtypes other than HER2-enriched. Furthermore, patients with TG levels increased after NAC in all subtypes, and the rate of change was highest especially in TNBC (21.0% → 48.1%).
Table 1
Differences in clinicopathological features due to intrinsic breast cancer subtypes in 327 patients
Parameters
Intrinsic subtype
P value
Luminal (n = 108)
Luminal-HER2 (n = 42)
HER2-enriched (n = 72)
TNBC (n = 105)
Age at operation
0.054
≤ 54
59 (54.6%)
24 (57.1%)
26 (36.1%)
55 (52.4%)
 
> 54
49 (45.4%)
18 (42.9%)
46 (63.9%)
50 (47.6%)
 
Menopause
0.006
Pre-
47 (43.5%)
22 (52.4%)
16 (22.9%)
36 (35.0%)
Post-
61 (56.5%)
20 (47.6%)
54 (77.1%)
67 (65.0%)
BMI
0.341
≤ 22.0
48 (44.4%)
21 (50.0%)
42 (58.3%)
53 (50.5%)
> 22.0
60 (55.6%)
21 (50.0%)
30 (41.7%)
52 (49.5%)
Tumor size
0.528
≤ 2 cm
13 (12.0%)
9 (21.4%)
9 (12.5%)
14 (13.3%)
> 2 cm
95 (88.0%)
33 (78.6%)
63 (87.5%)
91 (86.7%)
Lymph node status
0.011
Negative
27 (25.0%)
22 (52.4%)
26 (36.1%)
30 (28.6%)
Positive
81 (75.0%)
20 (47.6%)
46 (63.9%)
75 (71.4%)
TC (preNAC)
0.632
Normal
52 (54.2%)
21 (60.0%)
39 (60.9%)
49 (51.6%)
High
44 (45.8%)
14 (40.0%)
25 (39.1%)
46 (48.4%)
TG (preNAC)
0.999
Normal
76 (79.2%)
28 (77.8%)
48 (78.7%)
75 (79.0%)
High
20 (20.8%)
8 (22.2%)
13 (21.3%)
20 (21.0%)
TC (postNAC)
0.014
Normal
35 (37.6%)
15 (50.0%)
36 (62.1%)
33 (38.4%)
High
58 (62.4%)
15 (50.0%)
22 (37.9%)
53 (61.6%)
TG (postNAC)
0.649
Normal
53 (58.9%)
16 (55.2%)
33 (62.3%)
42 (51.9%)
High
37 (41.1%)
13 (44.8%)
20 (37.7%)
39 (48.1%)
Objective response rate
0.070
Non-ORR
11 (10.2%)
6 (14.3%)
2 (2.8%)
13 (12.4%)
ORR
97 (89.8%)
36 (85.7%)
70 (97.2%)
92 (87.6%)
Pathological response
< 0.001
Non-pCR
89 (82.4%)
30 (71.4%)
29 (40.3%)
58 (55.2%)
pCR
19 (17.6%)
12 (28.6%)
43 (59.7%)
47 (44.8%)
BMI, body mass index; HER2, human epidermal growth factor receptor 2; NAC, neoadjuvant chemotherapy; ORR, objective response rate; pCR, pathological complete response; TC, total-cholesterol; TG, triglyceride; TNBC, triple-negative breast cancer
In all breast cancer patients, RFS and OS were significantly longer in patients who achieved pCR than in those who did not (P < 0.001 and P = 0.006, log-rank, respectively; Additional file 1: Fig. S1a, Additional file 2: Fig. S2a). Furthermore, these outcomes were also significantly better in patients who achieved OR than in those who did not (P < 0.001 and P = 0.001, log-rank, respectively; Additional file 3: Figs. S3a, Additional file 4: Fig. S4a). In addition, we investigated the prognostic factors for RFS and OS for each breast cancer subtype. Among patients with luminal cancer, no significant differences were observed in RFS (P = 0.882, P = 0.399, log-rank, respectively) and OS (P = 0.861, P = 0.202, log-rank, respectively) according to the clinicopathological responses, pCR, and OR (Additional file 1: Fig. S1b, Additional file 2: Fig. S2b, Additional file 3: Fig. S3b, Additional file 4: Fig. S4b). In contrast, among the patients with TNBC, RFS (P = 0.005, P < 0.001, log-rank, respectively) and OS (P = 0.003, P < 0.001, log-rank, respectively) were significantly longer in patients who achieved pCR or OR than in those who did not (Additional file 1: Fig. S1e, Additional file 2: Fig. S2e, Additional file 3: Fig. S3e, Additional file 4: Fig. S4e).

Analysis of relationships between lipid metabolism and chemosensitivity and prognosis

The relationship between lipid metabolism and chemosensitivity was examined (Table 2). There were no significant correlations between lipid metabolism and the pCR in any breast cancer subtype. In contrast, only TNBC patients with OR had significantly higher TG levels after NAC than patients without OR (P = 0.049).
Table 2
Relationships between lipid metabolism and chemosensitivity
 
pCR
OR
Negative
Positive
P value
Negative
Positive
P value
All breast cancer (n = 327)
TC (preNAC)
0.626
  
0.242
 Normal
98 (54.1%)
63 (57.8%)
13 (44.8%)
148 (56.7%)
 High
83 (45.9%)
46 (42.2%)
16 (55.2%)
113 (43.3%)
TG (preNAC)
0.299
  
0.866
 Normal
139 (76.8%)
88 (82.2%)
24 (80.0%)
203 (78.7%)
 High
42 (23.2%)
19 (17.8%)
6 (20.0%)
55 (21.3%)
TC (postNAC)
0.703
  
0.952
 Normal
72 (43.4%)
47 (46.5%)
11 (44.0%)
108 (44.6%)
 High
94 (56.6%)
54 (53.5%)
14 (56.0%)
134 (55.4%)
TG (postNAC)
0.694
  
0.667
 Normal
88 (55.7%)
56 (59.0%)
15 (62.5%)
129 (56.3%)
 High
70 (44.3%)
39 (41.0%)
9 (37.5%)
100 (43.7%)
Luminal (n = 108)
TC (preNAC)
0.795
  
0.728
 Normal
43 (55.1%)
9 (50.0%)
4 (44.4%)
48 (55.2%)
 High
35 (44.9%)
9 (50.0%)
5 (55.6%)
39 (44.8%)
TG (preNAC)
0.347
  
0.680
 Normal
60 (76.9%)
16 (88.9%)
8 (88.9%)
68 (78.2%)
 High
18 (23.1%)
2 (11.1%)
1 (11.1%)
19 (21.8%)
TC (postNAC)
0.415
  
0.707
 Normal
27 (35.5%)
8 (47.1%)
2 (28.6%)
33 (38.4%)
 High
49 (64.5%)
9 (52.9%)
5 (71.4%)
53 (61.6%)
TG (postNAC)
0.785
  
0.685
 Normal
42 (57.5%)
11 (64.7%)
4 (66.7%)
49 (58.3%)
 High
31 (42.5%)
6 (35.3%)
2 (33.3%)
35 (41.7%)
Luminal-HER (n = 42)
TC (preNAC)
0.766
  
1.000
 Normal
14 (58.3%)
7 (63.6%)
3 (60.0%)
18 (60.0%)
 High
10 (41.7%)
4 (36.4%)
2 (40.0%)
12 (40.0%)
TG (preNAC)
0.076
  
0.109
 Normal
17 (68.0%)
11 (100.0%)
3 (50.0%)
25 (83.3%)
 High
8 (32.0%)
0 (0.0%)
3 (50.0%)
5 (16.7%)
TC (postNAC)
1.000
  
0.330
 Normal
10 (50.0%)
5 (50.0%)
1 (20.0%)
14 (56.0%)
 High
10 (50.0%)
5 (50.0%)
4 (80.0%)
11 (44.0%)
TG (postNAC)
0.130
  
0.144
 Normal
9 (45.0%)
7 (77.8%)
1 (20.0%)
15 (62.5%)
 High
11 (55.0%)
2 (22.2%)
4 (80.0%)
9 (37.5%)
HER2-enriched (n = 72)
TC (preNAC)
0.436
  
0.750
 Normal
14 (53.9%)
25 (65.8%)
1 (50.0%)
38 (61.3%)
 High
12 (46.1%)
13 (34.2%)
1 (50.0%)
24 (38.7%)
TG (preNAC)
0.835
  
0.323
 Normal
20 (80.0%)
28 (77.8%)
2 (100.0%)
46 (78.0%)
 High
5 (20.0%)
8 (22.2%)
0 (0.0%)
13 (22.0%)
TC (postNAC)
0.792
  
0.521
 Normal
15 (60.0%)
21 (63.6%)
2 (100.0%)
34 (60.7%)
 High
10 (40.0%)
12 (36.4%)
0 (0.0%)
22 (39.3%)
TG (postNAC)
0.779
  
0.719
 Normal
15 (65.2%)
18 (60.0%)
1 (50.0%)
32 (62.8%)
 High
8 (34.8%)
12 (40.0%)
1 (50.0%)
19 (37.2%)
TNBC (n = 105)
TC (preNAC)
0.889
  
0.378
 Normal
27 (50.9%)
22 (52.4%)
5 (38.5%)
44 (53.7%)
 High
26 (49.1%)
20 (47.6%)
8 (61.5%)
38 (46.3%)
TG (preNAC)
0.936
  
0.729
 Normal
42 (79.3%)
33 (78.6%)
11 (84.6%)
64 (78.1%)
 High
11 (20.7%)
9 (21.4%)
2 (15.4%)
18 (21.9%)
TC (postNAC)
0.270
  
0.322
 Normal
20 (44.5%)
13 (31.7%)
6 (54.6%)
27 (36.0%)
 High
25 (55.6%)
28 (68.3%)
5 (45.4%)
48 (64.0%)
TG (postNAC)
0.921
  
0.049
 Normal
22 (52.4%)
20 (51.3%)
9 (81.8%)
33 (47.1%)
 High
20 (47.6%)
19 (48.7%)
2 (18.2%)
37 (52.9%)
HER2, human epidermal growth factor receptor 2; NAC, neoadjuvant chemotherapy; TC, total-cholesterol; TG, triglyceride; TNBC, triple-negative breast cancer
We also investigated the prognostic value of serum lipid levels before and after NAC for each intrinsic breast cancer subtype. In patients with HER2-enriched breast cancer, those with normal TC levels before NAC had a significantly better OS than those with high TC levels (P = 0.013, log-rank test) (Fig. 1d), and in patients with TNBC, the group with high TC levels after NAC had significantly better OS than those with normal TC levels (P = 0.014, log-rank test) (Fig. 2e). There was no association between recurrence and TC levels. Also, there was no relationship between the prognosis and triglyceride levels before and after NAC (Additional files 510: Figs. S5–S10).

Analysis of relationships between lipid metabolism and immune activity

The pre-NAC ALC ranged from 712.8 to 4446.2 (mean, 1811.0; median, 1749; standard deviation, 613.9), and the pre-NAC NLR ranged from 0.5 to 10.6 (mean, 2.3; median, 2.0; standard deviation, 1.2). The post-NAC ALC ranged from 285.6 to 3697.7 (mean, 1122.4; median, 1005.4; standard deviation, 517.0), and the post-NAC NLR ranged from 0.3 to 15.9 (mean, 2.9; median, 2.4; standard deviation, 1.9). We defined the pre-NAC median as the cutoff value for the ALC and NLR. There were no significant correlations between the systemic immune activity and the effect of NAC in all the breast cancer patients or each of the breast cancer subtypes (Additional file 11: Table S1).
The relationship between lipid metabolism and systemic immune activity is shown in Table 3. Patients with a high ALC before NAC had significantly higher TG levels after NAC in all the breast cancers (P = 0.001). In addition, among the patients with HER2-enriched breast cancer, high TG levels after NAC were associated significantly with a high ALC before NAC (P = 0.021), and high TG levels before NAC were associated significantly with a high ALC after NAC (P = 0.046). Furthermore, among patients with TNBC, high TG levels after NAC were associated significantly with a high ALC (P = 0.008) and a low NLR (P = 0.025) before NAC, while high TG levels before NAC were associated significantly with a low NLR after NAC (P = 0.034).
Table 3
Relationships between lipid metabolism and immune activity
 
ALC (pre-NAC)
NLR (pre-NAC)
ALC (post-NAC)
NLR (post-NAC)
Low
High
P value
Low
High
P value
Low
High
P value
Low
High
P value
All breast cancer (n = 327)
TC (preNAC)
  
0.638
  
0.479
  
0.564
  
0.978
 Normal
82 (56.9%)
79 (54.1%)
 
76 (53.2%)
85 (57.8%)
 
145 (56.2%)
15 (50.0%)
 
59 (55.7%)
101 (55.5%)
 
 High
62 (43.1%)
67 (45.9%)
 
67 (46.8%)
62 (42.2%)
 
113 (43.8%)
15 (50.0%)
 
47 (44.3%)
81 (44.5%)
 
TG (preNAC)
  
0.249
  
0.886
  
0.101
  
0.655
 Normal
117 (81.8%)
110 (75.9%)
 
112 (79.4%)
115 (78.2%)
 
205 (80.1%)
20 (66.7%)
 
81 (77.1%)
144 (79.6%)
 
 High
26 (18.2%)
35 (24.1%)
 
29 (20.6%)
32 (21.8%)
 
51 (19.9%)
10 (33.3%)
 
24 (22.9%)
37 (20.4%)
 
TC (postNAC)
  
0.388
  
0.219
  
0.292
  
0.163
 Normal
60 (47.6%)
59 (41.8%)
 
60 (41.1%)
59 (48.8%)
 
105 (43.8%)
14 (56.0%)
 
51 (50.5%)
68 (41.5%)
 
 High
66 (52.4%)
82 (58.2%)
 
86 (58.9%)
62 (51.2%)
 
135 (56.2%)
11 (44.0%)
 
50 (49.5%)
96 (58.5%)
 
TG (postNAC)
  
0.001
  
0.164
  
0.803
  
0.974
 Normal
81 (68.6%)
63 (46.7%)
 
73 (52.9%)
71 (61.7%)
 
129 (56.8%)
13 (54.2%)
 
55 (56.7%)
87 (56.5%)
 
 High
37 (31.4%)
72 (53.3%)
 
65 (47.1%)
44 (38.3%)
 
98 (43.2%)
11 (45.8%)
 
42 (43.3%)
67 (43.5%)
 
Luminal (n = 108)
TC (preNAC)
  
0.54
  
0.838
  
0.728
  
0.665
 Normal
25 (58.1%)
27 (50.9%)
 
28 (52.8%)
24 (55.8%)
 
48 (55.2%)
4 (44.4%)
 
16 (50.0%)
36 (56.3%)
 
 High
18 (41.9%)
26 (49.1%)
 
25 (47.2%)
19 (44.2%)
 
39 (44.8%)
5 (55.6%)
 
16 (50.0%)
28 (43.7%)
 
TG (preNAC)
  
0.801
  
0.801
  
0.915
  
0.064
 Normal
35 (81.4%)
41 (77.4%)
 
41 (77.4%)
35 (81.4%)
 
69 (79.3%)
7 (77.8%)
 
29 (90.6%)
47 (73.4%)
 
 High
8 (18.6%)
12 (22.6%)
 
12 (22.6%)
8 (18.6%)
 
18 (20.7%)
2 (22.2%)
 
3 (9.4%)
17 (26.6%)
 
TC (postNAC)
  
0.853
  
0.667
  
0.926
  
0.38
 Normal
15 (36.6%)
20 (38.5%)
 
20 (35.7%)
15 (40.5%)
 
31 (37.8%)
4 (36.4%)
 
16 (44.4%)
19 (33.3%)
 
 High
26 (63.4%)
32 (61.5%)
 
36 (64.3%)
22 (59.5%)
 
51 (62.2%)
7 (63.6%)
 
20 (55.6%)
38 (66.7%)
 
TG (postNAC)
  
0.397
  
0.386
  
0.515
  
0.661
 Normal
25 (64.1%)
28 (54.9%)
 
34 (63.0%)
19 (52.8%)
 
45 (57.0%)
8 (72.7%)
 
22 (62.9%)
31 (56.4%)
 
 High
14 (35.9%)
23 (45.1%)
 
20 (37.0%)
17 (47.2%)
 
34 (43.0%)
3 (27.3%)
 
13 (37.1%)
24 (43.6%)
 
Luminal-HER (n = 42)
TC (preNAC)
  
0.176
  
0.491
  
0.135
  
0.774
 Normal
13 (72.2%)
8 (47.1%)
 
10 (52.6%)
11 (68.8%)
 
19 (65.5%)
1 (20.0%)
 
9 (56.3%)
11 (61.1%)
 
 High
5 (27.8%)
9 (52.9%)
 
9 (47.4%)
5 (31.2%)
 
10 (34.5%)
4 (80.0%)
 
7 (43.7%)
7 (38.9%)
 
TG (preNAC)
  
0.695
  
0.114
  
0.868
  
0.7
 Normal
14 (73.7%)
14 (82.4%)
 
17 (89.5%)
11 (64.7%)
 
23 (76.7%)
4 (80.0%)
 
13 (81.3%)
14 (73.7%)
 
 High
5 (26.3%)
3 (17.6%)
 
2 (10.5%)
6 (35.3%)
 
7 (23.3%)
1 (20.0%)
 
3 (18.7%)
5 (26.3%)
 
TC (postNAC)
  
0.462
  
0.715
  
0.96
  
0.837
 Normal
10 (58.8%)
5 (38.5%)
 
8 (57.1%)
7 (43.8%)
 
14 (51.9%)
1 (50.0%)
 
7 (53.9%)
8 (50.0%)
 
 High
7 (41.2%)
8 (61.5%)
 
6 (42.9%)
9 (56.2%)
 
13 (48.1%)
1 (50.0%)
 
6 (46.1%)
8 (50.0%)
 
TG (postNAC)
  
0.274
  
0.897
  
0.206
  
0.705
 Normal
11 (64.7%)
5 (41.7%)
 
7 (53.9%)
9 (56.3%)
 
15 (57.7%)
0 (0.00%)
 
6 (46.2%)
9 (60.0%)
 
 High
6 (35.3%)
7 (58.3%)
 
6 (46.1%)
7 (43.7%)
 
11 (42.3%)
2 (100.0%)
 
7 (53.8%)
6 (40.0%)
 
HER2-enriched (n = 72)
TC (preNAC)
  
0.955
  
0.935
  
0.738
  
0.935
 Normal
20 (60.6%)
19 (61.3%)
 
16 (61.5%)
23 (60.5%)
 
33 (62.3%)
6 (54.6%)
 
16 (61.5%)
23 (60.5%)
 
 High
13 (39.4%)
12 (38.7%)
 
10 (38.5%)
15 (39.5%)
 
20 (37.7%)
5 (45.4%)
 
10 (38.5%)
15 (39.5%)
 
TG (preNAC)
  
0.363
  
0.539
  
0.046
  
0.349
 Normal
26 (83.9%)
22 (73.3%)
 
20 (83.3%)
28 (75.7%)
 
42 (84.0%)
6 (54.6%)
 
18 (72.0%)
30 (83.3%)
 
 High
5 (16.1%)
8 (26.7%)
 
4 (16.7%)
9 (24.3%)
 
8 (16.0%)
5 (45.4%)
 
7 (28.0%)
6 (16.7%)
 
TC (postNAC)
  
0.593
  
0.787
  
0.697
  
0.267
 Normal
16 (66.7%)
20 (58.8%)
 
17 (58.6%)
19 (65.5%)
 
30 (60.0%)
6 (75.0%)
 
16 (72.7%)
20 (55.6%)
 
 High
8 (33.3%)
14 (41.2%)
 
12 (41.4%)
10 (34.5%)
 
20 (40.0%)
2 (25.0%)
 
6 (27.3%)
16 (44.4%)
 
TG (postNAC)
  
0.021
  
0.264
  
0.766
  
0.779
 Normal
18 (81.8%)
15 (48.4%)
 
14 (53.9%)
19 (70.4%)
 
29 (63.0%)
4 (57.1%)
 
13 (65.0%)
20 (60.6%)
 
 High
4 (18.2%)
16 (51.6%)
 
12 (46.1%)
8 (29.6%)
 
17 (37.0%)
3 (42.9%)
 
7 (35.0%)
13 (39.4%)
 
TNBC (n = 105)
TC (preNAC)
  
0.539
  
0.683
  
0.364
  
0.664
 Normal
24 (48.0%)
25 (55.6%)
 
22 (48.9%)
27 (54.0%)
 
45 (50.6%)
4 (80.0%)
 
18 (56.3%)
31 (50.0%)
 
 High
26 (52.0%)
20 (44.4%)
 
23 (51.1%)
23 (46.0%)
 
44 (49.4%)
1 (20.0%)
 
14 (43.7%)
31 (50.0%)
 
TG (preNAC)
  
0.22
  
0.462
  
0.287
  
0.034
 Normal
42 (84.0%)
33 (73.3%)
 
34 (75.6%)
41 (82.0%)
 
71 (79.8%)
3 (60.0%)
 
21 (65.6%)
53 (85.5%)
 
 High
8 (16.0%)
12 (26.7%)
 
11 (24.4%)
9 (18.0%)
 
18 (20.2%)
2 (40.0%)
 
11 (34.4%)
9 (14.5%)
 
TC (postNAC)
  
0.382
  
0.19
  
0.294
  
0.87
 Normal
19 (43.2%)
14 (33.3%)
 
15 (31.9%)
18 (46.2%)
 
30 (37.0%)
3 (75.0%)
 
12 (40.0%)
21 (38.2%)
 
 High
25 (56.8%)
28 (66.7%)
 
32 (68.1%)
21 (53.8%)
 
51 (63.0%)
1 (25.0%)
 
18 (60.0%)
34 (61.8%)
 
TG (postNAC)
  
0.008
  
0.025
  
0.353
  
0.817
 Normal
27 (67.5%)
15 (36.6%)
 
18 (40.0%)
24 (66.7%)
40 (52.6%)
1 (25.0%)
14 (48.3%)
27 (52.9%)
 
 High
13 (32.5%)
26 (63.4%)
 
27 (60.0%)
12 (33.3%)
36 (47.4%)
3 (75.0%)
15 (51.7%)
24 (47.1%)
 
ALC, absolute lymphocyte count; HER2, human epidermal growth factor receptor 2; NAC, neoadjuvant chemotherapy; NLR, neutrophil-to-lymphocyte ratio; TC, total-cholesterol; TG, triglyceride; TNBC, triple-negative breast cancer

Discussion

In the present study, NAC increased serum TG levels, particularly in patients with TNBC. Some previous studies showed that serum lipid levels increased significantly after chemotherapy and that the TG levels may be a sensitive biomarker for determining the effect of adjuvant chemotherapy [24, 30]. Many anticancer drugs are metabolized in liver and may cause non-alcoholic fatty liver disease by variety of mechanisms [31]. However, this phenomenon has not yet been fully studied. To the best of our knowledge, our study was the first to analyze the predictive value of lipid metabolism for chemosensitivity of breast cancer patients treated with NAC and to stratify the intrinsic subtypes of breast cancer.
In this study, patients with reduced tumor size had significantly higher TG levels after NAC in only TNBC. Sharma et al. reported that some chemotherapy agents affect serum lipid levels by regulating the expression of genes involved in lipid metabolism in liver cells [32]. Therefore, it is considered that there is a correlation between the effects of NAC and lipid metabolism in TNBC.
The efficacy of NAC, especially in terms of the pCR, is currently acknowledged as an indicator of good outcomes in patients with TNBC and HER2-enriched breast cancer, which have high immune activity [3, 33, 34]. Hence, it is expected that there will be an association between lipid metabolism and tumor immune activity in TNBC. Recent studies have reported that the regulation of metabolism can affect the tumor immune microenvironment and enhance the antitumor immune response [57]. In our study, good systemic immune activity, a high ALC, or low NLR before NAC were associated significantly with high TG levels after NAC in patients with TNBC or HER2-enriched breast cancer.
However, no relationships were observed between the pre-NAC lipid levels and NAC efficacy. In addition, the serum lipid levels before NAC showed no significant relationships with the ALC or NLR. Hence, it was difficult to predict chemosensitivity or systemic immune activity based on serum lipid levels prior to NAC.
In the present study, no significant associations were observed between the systemic immune activity and the effect of NAC. However, in our previous study, we set the cutoff value of pre-NAC NLR to 3.0, in the same breast cancer patients, and the pCR rate was significantly higher in TNBC patients with a good immune status, low NLR group [4]. This result suggested that not only the effect of tumor reduction, but also the effect of increasing serum lipid levels is recognized in patients with good systemic immune activity.
Although the TG levels after NAC may be an indicator of chemosensitivity in TNBC, they are not useful predictive markers of recurrence. The reason for this may be that changes in the lipid profiles after NAC are temporary [26]. We presumed that a favorable prognosis may not be based on lipid levels at the time of diagnosis or after NAC, but is induced by the maintenance good lipid metabolism after surgery.
This study has some limitations. First, this was a single-center, retrospective study, then the sample size was relatively small. Second, in our study, serum TC levels were associated with better OS in patients with HER2-enriched breast cancer or TNBC. However, we did not have detailed data on high-density lipoprotein cholesterol and low-density lipoprotein cholesterol levels. In addition, many factors influence serum lipid levels, including lifestyle and adherence to medication. Considering these limitations, further prospective multicenter studies are needed.

Conclusions

This is the first study to demonstrate the clinical relationships between lipid metabolism, chemosensitivity, and systemic immune activity in patients with breast cancer treated with NAC. The findings of this study indicated that a good systemic immune activity and the effect of NAC may be associated with lipid metabolism regulated by chemotherapy in patients with TNBC.

Acknowledgements

We thank Tomomi Okawa (Department of Breast Surgical Oncology, Osaka Metropolitan University Graduate School of Medicine) for helpful advice regarding data management.

Declarations

A written informed consent to participate in the study was obtained from each subject in accordance with the declaration of Helsinki principles. Each patient or the patient’s family was fully 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 (approve number #926).
Not applicable.

Competing interests

The authors declare that they have no competing interests.
Open AccessThis article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://​creativecommons.​org/​licenses/​by/​4.​0/​. 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 in a credit line to the data.

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Anhänge

Supplementary Information

Literatur
1.
Zurück zum Zitat Chen AM, Meric-Bernstam F, Hunt KK, Thames HD, Oswald MJ, Outlaw ED, Strom EA, McNeese MD, Kuerer HM, Ross MI, et al. Breast conservation after neoadjuvant chemotherapy: the MD Anderson cancer center experience. J Clin Oncol. 2004;22(12):2303–12.CrossRef Chen AM, Meric-Bernstam F, Hunt KK, Thames HD, Oswald MJ, Outlaw ED, Strom EA, McNeese MD, Kuerer HM, Ross MI, et al. Breast conservation after neoadjuvant chemotherapy: the MD Anderson cancer center experience. J Clin Oncol. 2004;22(12):2303–12.CrossRef
2.
Zurück zum Zitat Beriwal S, Schwartz GF, Komarnicky L, Garcia-Young JA. Breast-conserving therapy after neoadjuvant chemotherapy: long-term results. Breast J. 2006;12(2):159–64.CrossRef Beriwal S, Schwartz GF, Komarnicky L, Garcia-Young JA. Breast-conserving therapy after neoadjuvant chemotherapy: long-term results. Breast J. 2006;12(2):159–64.CrossRef
3.
Zurück zum Zitat Cortazar P, Zhang L, Untch M, Mehta K, Costantino JP, Wolmark N, Bonnefoi H, Cameron D, Gianni L, Valagussa P, et al. Pathological complete response and long-term clinical benefit in breast cancer: the CTNeoBC pooled analysis. Lancet. 2014;384(9938):164–72.CrossRef Cortazar P, Zhang L, Untch M, Mehta K, Costantino JP, Wolmark N, Bonnefoi H, Cameron D, Gianni L, Valagussa P, et al. Pathological complete response and long-term clinical benefit in breast cancer: the CTNeoBC pooled analysis. Lancet. 2014;384(9938):164–72.CrossRef
4.
Zurück zum Zitat Asano Y, Kashiwagi S, Onoda N, Noda S, Kawajiri H, Takashima T, Ohsawa M, Kitagawa S, Hirakawa K. Predictive value of neutrophil/lymphocyte ratio for efficacy of preoperative chemotherapy in triple-negative breast cancer. Ann Surg Oncol. 2016;23(4):1104–10.CrossRef Asano Y, Kashiwagi S, Onoda N, Noda S, Kawajiri H, Takashima T, Ohsawa M, Kitagawa S, Hirakawa K. Predictive value of neutrophil/lymphocyte ratio for efficacy of preoperative chemotherapy in triple-negative breast cancer. Ann Surg Oncol. 2016;23(4):1104–10.CrossRef
5.
Zurück zum Zitat Ho PC, Liu PS. Metabolic communication in tumors: a new layer of immunoregulation for immune evasion. J Immunother Cancer. 2016;4:4.CrossRef Ho PC, Liu PS. Metabolic communication in tumors: a new layer of immunoregulation for immune evasion. J Immunother Cancer. 2016;4:4.CrossRef
6.
Zurück zum Zitat Allison KE, Coomber BL, Bridle BW. Metabolic reprogramming in the tumour microenvironment: a hallmark shared by cancer cells and T lymphocytes. Immunology. 2017;152(2):175–84.CrossRef Allison KE, Coomber BL, Bridle BW. Metabolic reprogramming in the tumour microenvironment: a hallmark shared by cancer cells and T lymphocytes. Immunology. 2017;152(2):175–84.CrossRef
7.
Zurück zum Zitat Li X, Wenes M, Romero P, Huang SC, Fendt SM, Ho PC. Navigating metabolic pathways to enhance antitumour immunity and immunotherapy. Nat Rev Clin Oncol. 2019;16(7):425–41.CrossRef Li X, Wenes M, Romero P, Huang SC, Fendt SM, Ho PC. Navigating metabolic pathways to enhance antitumour immunity and immunotherapy. Nat Rev Clin Oncol. 2019;16(7):425–41.CrossRef
8.
Zurück zum Zitat Renehan AG, Tyson M, Egger M, Heller RF, Zwahlen M. Body-mass index and incidence of cancer: a systematic review and meta-analysis of prospective observational studies. Lancet. 2008;371(9612):569–78.CrossRef Renehan AG, Tyson M, Egger M, Heller RF, Zwahlen M. Body-mass index and incidence of cancer: a systematic review and meta-analysis of prospective observational studies. Lancet. 2008;371(9612):569–78.CrossRef
9.
Zurück zum Zitat Wada K, Nagata C, Tamakoshi A, Matsuo K, Oze I, Wakai K, Tsuji I, Sugawara Y, Mizoue T, Tanaka K, et al. Body mass index and breast cancer risk in Japan: a pooled analysis of eight population-based cohort studies. Ann Oncol. 2014;25(2):519–24.CrossRef Wada K, Nagata C, Tamakoshi A, Matsuo K, Oze I, Wakai K, Tsuji I, Sugawara Y, Mizoue T, Tanaka K, et al. Body mass index and breast cancer risk in Japan: a pooled analysis of eight population-based cohort studies. Ann Oncol. 2014;25(2):519–24.CrossRef
10.
Zurück zum Zitat Ahern TP, Lash TL, Damkier P, Christiansen PM, Cronin-Fenton DP. Statins and breast cancer prognosis: evidence and opportunities. Lancet Oncol. 2014;15(10):e461-468.CrossRef Ahern TP, Lash TL, Damkier P, Christiansen PM, Cronin-Fenton DP. Statins and breast cancer prognosis: evidence and opportunities. Lancet Oncol. 2014;15(10):e461-468.CrossRef
11.
Zurück zum Zitat Yu X, Luo Y, Zhou Y, Zhang Q, Wang J, Wei N, Mi M, Zhu J, Wang B, Chang H, et al. BRCA1 overexpression sensitizes cancer cells to lovastatin via regulation of cyclin D1-CDK4-p21WAF1/CIP1 pathway: analyses using a breast cancer cell line and tumoral xenograft model. Int J Oncol. 2008;33(3):555–63. Yu X, Luo Y, Zhou Y, Zhang Q, Wang J, Wei N, Mi M, Zhu J, Wang B, Chang H, et al. BRCA1 overexpression sensitizes cancer cells to lovastatin via regulation of cyclin D1-CDK4-p21WAF1/CIP1 pathway: analyses using a breast cancer cell line and tumoral xenograft model. Int J Oncol. 2008;33(3):555–63.
12.
Zurück zum Zitat Wang G, Cao R, Wang Y, Qian G, Dan HC, Jiang W, Ju L, Wu M, Xiao Y, Wang X. Simvastatin induces cell cycle arrest and inhibits proliferation of bladder cancer cells via PPARgamma signalling pathway. Sci Rep. 2016;6:35783.CrossRef Wang G, Cao R, Wang Y, Qian G, Dan HC, Jiang W, Ju L, Wu M, Xiao Y, Wang X. Simvastatin induces cell cycle arrest and inhibits proliferation of bladder cancer cells via PPARgamma signalling pathway. Sci Rep. 2016;6:35783.CrossRef
13.
Zurück zum Zitat Weis M, Heeschen C, Glassford AJ, Cooke JP. Statins have biphasic effects on angiogenesis. Circulation. 2002;105(6):739–45.CrossRef Weis M, Heeschen C, Glassford AJ, Cooke JP. Statins have biphasic effects on angiogenesis. Circulation. 2002;105(6):739–45.CrossRef
14.
Zurück zum Zitat Massaro M, Zampolli A, Scoditti E, Carluccio MA, Storelli C, Distante A, De Caterina R. Statins inhibit cyclooxygenase-2 and matrix metalloproteinase-9 in human endothelial cells: anti-angiogenic actions possibly contributing to plaque stability. Cardiovasc Res. 2010;86(2):311–20.CrossRef Massaro M, Zampolli A, Scoditti E, Carluccio MA, Storelli C, Distante A, De Caterina R. Statins inhibit cyclooxygenase-2 and matrix metalloproteinase-9 in human endothelial cells: anti-angiogenic actions possibly contributing to plaque stability. Cardiovasc Res. 2010;86(2):311–20.CrossRef
15.
Zurück zum Zitat Mandal CC, Ghosh-Choudhury N, Yoneda T, Choudhury GG, Ghosh-Choudhury N. Simvastatin prevents skeletal metastasis of breast cancer by an antagonistic interplay between p53 and CD44. J Biol Chem. 2011;286(13):11314–27.CrossRef Mandal CC, Ghosh-Choudhury N, Yoneda T, Choudhury GG, Ghosh-Choudhury N. Simvastatin prevents skeletal metastasis of breast cancer by an antagonistic interplay between p53 and CD44. J Biol Chem. 2011;286(13):11314–27.CrossRef
16.
Zurück zum Zitat Beckwitt CH, Clark AM, Ma B, Whaley D, Oltvai ZN, Wells A. Statins attenuate outgrowth of breast cancer metastases. Br J Cancer. 2018;119(9):1094–105.CrossRef Beckwitt CH, Clark AM, Ma B, Whaley D, Oltvai ZN, Wells A. Statins attenuate outgrowth of breast cancer metastases. Br J Cancer. 2018;119(9):1094–105.CrossRef
17.
Zurück zum Zitat Crocetto F, Pandolfo SD, Aveta A, Martino R, Trama F, Caputo VF, Barone B, Abate M, Sicignano E, Cilio S et al. A comparative study of the triglycerides/HDL ratio and pseudocholinesterase levels in patients with bladder cancer. Diagnostics (Basel). 2022;12(2). Crocetto F, Pandolfo SD, Aveta A, Martino R, Trama F, Caputo VF, Barone B, Abate M, Sicignano E, Cilio S et al. A comparative study of the triglycerides/HDL ratio and pseudocholinesterase levels in patients with bladder cancer. Diagnostics (Basel). 2022;12(2).
18.
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
19.
Zurück zum Zitat Vacchelli E, Galluzzi L, Fridman WH, Galon J, Sautes-Fridman C, Tartour E, Kroemer G. Trial watch: chemotherapy with immunogenic cell death inducers. Oncoimmunology. 2012;1(2):179–88.CrossRef Vacchelli E, Galluzzi L, Fridman WH, Galon J, Sautes-Fridman C, Tartour E, Kroemer G. Trial watch: chemotherapy with immunogenic cell death inducers. Oncoimmunology. 2012;1(2):179–88.CrossRef
20.
Zurück zum Zitat Ladoire S, Arnould L, Apetoh L, Coudert B, Martin F, Chauffert B, Fumoleau P, Ghiringhelli F. Pathologic complete response to neoadjuvant chemotherapy of breast carcinoma is associated with the disappearance of tumor-infiltrating foxp3+ regulatory T cells. Clin Cancer Res. 2008;14(8):2413–20.CrossRef Ladoire S, Arnould L, Apetoh L, Coudert B, Martin F, Chauffert B, Fumoleau P, Ghiringhelli F. Pathologic complete response to neoadjuvant chemotherapy of breast carcinoma is associated with the disappearance of tumor-infiltrating foxp3+ regulatory T cells. Clin Cancer Res. 2008;14(8):2413–20.CrossRef
21.
Zurück zum Zitat Miyashita M, Sasano H, Tamaki K, Chan M, Hirakawa H, Suzuki A, Tada H, Watanabe G, Nemoto N, Nakagawa S, et al. Tumor-infiltrating CD8+ and FOXP3+ lymphocytes in triple-negative breast cancer: its correlation with pathological complete response to neoadjuvant chemotherapy. Breast Cancer Res Treat. 2014;148(3):525–34.CrossRef Miyashita M, Sasano H, Tamaki K, Chan M, Hirakawa H, Suzuki A, Tada H, Watanabe G, Nemoto N, Nakagawa S, et al. Tumor-infiltrating CD8+ and FOXP3+ lymphocytes in triple-negative breast cancer: its correlation with pathological complete response to neoadjuvant chemotherapy. Breast Cancer Res Treat. 2014;148(3):525–34.CrossRef
22.
Zurück zum Zitat Asano Y, Kashiwagi S, Goto W, Kurata K, Noda S, Takashima T, Onoda N, Tanaka S, Ohsawa M, Hirakawa K. Tumour-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. Tumour-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
23.
Zurück zum Zitat Goto W, Kashiwagi S, Takada K, Asano Y, Takahashi K, Fujita H, Takashima T, Tomita S, Hirakawa K, Ohira M. Significance of intrinsic breast cancer subtypes on the long-term prognosis after neoadjuvant chemotherapy. J Transl Med. 2018;16(1):307.CrossRef Goto W, Kashiwagi S, Takada K, Asano Y, Takahashi K, Fujita H, Takashima T, Tomita S, Hirakawa K, Ohira M. Significance of intrinsic breast cancer subtypes on the long-term prognosis after neoadjuvant chemotherapy. J Transl Med. 2018;16(1):307.CrossRef
24.
Zurück zum Zitat Alexopoulos CG, Pournaras S, Vaslamatzis M, Avgerinos A, Raptis S. Changes in serum lipids and lipoproteins in cancer patients during chemotherapy. Cancer Chemother Pharmacol. 1992;30(5):412–6.CrossRef Alexopoulos CG, Pournaras S, Vaslamatzis M, Avgerinos A, Raptis S. Changes in serum lipids and lipoproteins in cancer patients during chemotherapy. Cancer Chemother Pharmacol. 1992;30(5):412–6.CrossRef
25.
Zurück zum Zitat Arpino G, De Angelis C, Buono G, Colao A, Giuliano M, Malgieri S, Cicala S, De Laurentiis M, Accurso A, Crispo A, et al. Metabolic and anthropometric changes in early breast cancer patients receiving adjuvant therapy. Breast Cancer Res Treat. 2015;154(1):127–32.CrossRef Arpino G, De Angelis C, Buono G, Colao A, Giuliano M, Malgieri S, Cicala S, De Laurentiis M, Accurso A, Crispo A, et al. Metabolic and anthropometric changes in early breast cancer patients receiving adjuvant therapy. Breast Cancer Res Treat. 2015;154(1):127–32.CrossRef
26.
Zurück zum Zitat Tian W, Yao Y, Fan G, Zhou Y, Wu M, Xu D, Deng Y. Changes in lipid profiles during and after (neo)adjuvant chemotherapy in women with early-stage breast cancer: a retrospective study. PLoS ONE. 2019;14(8): e0221866.CrossRef Tian W, Yao Y, Fan G, Zhou Y, Wu M, Xu D, Deng Y. Changes in lipid profiles during and after (neo)adjuvant chemotherapy in women with early-stage breast cancer: a retrospective study. PLoS ONE. 2019;14(8): e0221866.CrossRef
27.
Zurück zum Zitat Greene FL, Sobin LH. A worldwide approach to the TNM staging system: collaborative efforts of the AJCC and UICC. J Surg Oncol. 2009;99(5):269–72.CrossRef Greene FL, Sobin LH. A worldwide approach to the TNM staging system: collaborative efforts of the AJCC and UICC. J Surg Oncol. 2009;99(5):269–72.CrossRef
28.
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 tumours: revised RECIST guideline (version 1.1). Eur J Cancer. 2009’45(2):228–47. 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 tumours: revised RECIST guideline (version 1.1). Eur J Cancer. 2009’45(2):228–47.
29.
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
30.
Zurück zum Zitat Rzymowska J. Effect of cytotoxic chemotherapy on serum lipid levels in breast cancer patients. Pathobiology. 1999;67(3):129–32.CrossRef Rzymowska J. Effect of cytotoxic chemotherapy on serum lipid levels in breast cancer patients. Pathobiology. 1999;67(3):129–32.CrossRef
31.
Zurück zum Zitat Tarantino G, Citro V, Capone D: Nonalcoholic fatty liver disease: a challenge from mechanisms to therapy. J Clin Med. 2019;9(1). Tarantino G, Citro V, Capone D: Nonalcoholic fatty liver disease: a challenge from mechanisms to therapy. J Clin Med. 2019;9(1).
32.
Zurück zum Zitat Sharma AK, Wen L, Hall LR, Allan JG, Clark BJ. Metabolism of pyrithiobac sodium in soils and sediment, addressing bound residues via kinetics modeling. J Agric Food Chem. 2016;64(29):5793–802.CrossRef Sharma AK, Wen L, Hall LR, Allan JG, Clark BJ. Metabolism of pyrithiobac sodium in soils and sediment, addressing bound residues via kinetics modeling. J Agric Food Chem. 2016;64(29):5793–802.CrossRef
33.
Zurück zum Zitat von Minckwitz G, Untch M, Blohmer JU, Costa SD, Eidtmann H, Fasching PA, Gerber B, Eiermann W, Hilfrich J, Huober J, et al. Definition and impact of pathologic complete response on prognosis after neoadjuvant chemotherapy in various intrinsic breast cancer subtypes. J Clin Oncol. 2012;30(15):1796–804.CrossRef von Minckwitz G, Untch M, Blohmer JU, Costa SD, Eidtmann H, Fasching PA, Gerber B, Eiermann W, Hilfrich J, Huober J, et al. Definition and impact of pathologic complete response on prognosis after neoadjuvant chemotherapy in various intrinsic breast cancer subtypes. J Clin Oncol. 2012;30(15):1796–804.CrossRef
34.
Zurück zum Zitat Bonnefoi H, Litiere S, Piccart M, MacGrogan G, Fumoleau P, Brain E, Petit T, Rouanet P, Jassem J, Moldovan C, et al. Pathological complete response after neoadjuvant chemotherapy is an independent predictive factor irrespective of simplified breast cancer intrinsic subtypes: a landmark and two-step approach analyses from the EORTC 10994/BIG 1–00 phase III trial. Ann Oncol. 2014;25(6):1128–36.CrossRef Bonnefoi H, Litiere S, Piccart M, MacGrogan G, Fumoleau P, Brain E, Petit T, Rouanet P, Jassem J, Moldovan C, et al. Pathological complete response after neoadjuvant chemotherapy is an independent predictive factor irrespective of simplified breast cancer intrinsic subtypes: a landmark and two-step approach analyses from the EORTC 10994/BIG 1–00 phase III trial. Ann Oncol. 2014;25(6):1128–36.CrossRef
Metadaten
Titel
Clinical verification of the relationship between serum lipid metabolism and immune activity in breast cancer patients treated with neoadjuvant chemotherapy
verfasst von
Wataru Goto
Shinichiro Kashiwagi
Koji Takada
Yuka Asano
Kana Ogisawa
Tamami Morisaki
Masatsune Shibutani
Hiroaki Tanaka
Kiyoshi Maeda
Publikationsdatum
01.12.2023
Verlag
BioMed Central
Erschienen in
European Journal of Medical Research / Ausgabe 1/2023
Elektronische ISSN: 2047-783X
DOI
https://doi.org/10.1186/s40001-022-00964-w

Weitere Artikel der Ausgabe 1/2023

European Journal of Medical Research 1/2023 Zur Ausgabe