Skip to main content
Erschienen in: World Journal of Surgical Oncology 1/2020

Open Access 01.12.2020 | Research

HIF-1α expression in liver metastasis but not primary colorectal cancer is associated with prognosis of patients with colorectal liver metastasis

verfasst von: Yuma Wada, Yuji Morine, Satoru Imura, Tetsuya Ikemoto, Yu Saito, Chie Takasu, Shinichiro Yamada, Mitsuo Shimada

Erschienen in: World Journal of Surgical Oncology | Ausgabe 1/2020

Abstract

Background

The role of hypoxia-inducible factor-1α (HIF-1α) in primary colorectal cancer (CRC) and colorectal liver metastasis (CRLM) has remained unclear. The aim of this study was to investigate HIF-1α expression and its association with prognosis in patients with CRLM with a focus on hepatic stellate cells (HSCs).

Methods

Colon cancer cells were cultured in HSC-conditioned medium (CM), and HIF-1α expression and cell migration were analyzed. Seventy-five patients with CRLM who underwent an initial curative hepatectomy were enrolled. We examined HIF-1α expressions and patient prognosis between primary CRCs and the matched liver metastatic specimens.

Results

Activated HSCs induced HIF-1α mRNA and protein expression in colon cancer cells (p < 0.01) and promoted cell migration (p < 0.01). The positive rates of HIF-1α expression in primary CRCs and liver metastases were 68.0 and 72.0%, respectively. There were no differences in overall (OS) and disease-free survival (DFS) of HIF-1α expression in primary CRC. However, HIF-1α expression in liver metastasis correlated to poor prognosis in both OS and DFS. Furthermore, patients with HIF-1α positive expression in liver metastasis had poor prognosis.

Conclusion

HIF-1α expression in liver metastasis determines poor prognosis of CRLM patients. HSCs might play a key role in aggressive phenotypes of tumor cells.
Hinweise

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Abkürzungen
CRC
Colorectal cancer
CRLM
Colorectal liver metastasis
HSC
Hepatic stellate cell
CAF
Cancer-associated fibroblast
HIF-1α
Hypoxia-inducible factor-1α
OS
Overall survival
DFS
Disease-free survival
TAM
Tumor-associated macrophage
EMT
Epithelial mesenchymal transition
CM
Conditioned medium
IL-6
Interleukin-6

Background

In patients with colorectal liver metastasis (CRLM), surgical resection improves patient prognosis and is recommended if the resection will be curative [15]. On the other hand, with the implementation of “targeted” molecular therapies against epidermal growth factor receptor (EGFR) and vascular endothelial growth factor (VEGF), the median overall survival (OS) of patients with metastatic colorectal cancer (CRC) has progressively improved, surpassing 30 months [68]. The monoclonal antibodies like cetuximab, panitumumab, and bevacizumab have demonstrated effectiveness, both in terms of better response and improved survival. A comparison of various markers between primary and metastatic tumors have been recently reported [913]. Previous studies reported that while the immune microenvironment in the primary CRC tumor and liver metastasis is different, hypoxia-inducible factor-1α (HIF-1α) expression in primary CRC was comparable to that in corresponding metastases and HIF-1α expression is consistent in primary CRC and matched metastatic tissues [9, 10]. However, whether primary CRC and the associated metastases have similar molecular features remains unknown. A comparative analysis between primary CRC and metastatic tumors may improve understanding of the various molecular alterations in metastatic tumors and facilitate research and development of novel targeted drugs for CRC.
Once cancer cells in the primary tumor site migrate to distant metastatic sites, some cells around the tumor cells, such as sinusoidal endothelial cells, macrophages, or fibroblasts, can increase tumor malignancy [14]. In the liver microenvironment, hepatocytes, Kupffer cells, and hepatic stellate cells (HSCs) play an important role [1517]. In the liver cancer microenvironment, Kupffer cells and HSCs are activated by cancer cells, and tumor-associated macrophages (TAMs) and activated HSCs regulate tumor malignant behavior [17]. HSCs play a key role in the development of aggressive phenotypes of tumor cells. We previously reported that activated HSCs promoted cancer cell progression through paracrine or autocrine interleukin-6 (IL-6) [18]. However, there are few reports about the relationship between activated HSCs, which are considered cancer-associated fibroblasts, and metastatic cancer cells in CRLM. We speculate that the characteristics of metastatic cancer cells could be modified by cancer-associated fibroblasts in the cancer microenvironment of liver.
The HIF-1α transcription factor [1923] plays a central role in biologic processes under hypoxic conditions including angiogenesis [24, 25], tumor growth [26], and epithelial mesenchymal transition [27] in several cancer types. A previous report showed that high HIF-1α expression correlated to tumor malignancy in liver compared with some metastatic organs such as bone and lung [28]. Moreover, another report showed that HIF-1α expression was altered from primary sites to metastatic sites, and high expression of HIF-1α in the metastatic site correlated to poor prognosis [29]. Therefore, HIF-1α expression might serve a critical role to regulate tumor malignancy in CRLM.
The aim of this study was to elucidate a possible mechanism of activated HSCs on augmenting tumor malignancy and to investigate the association of HIF-1α expression between primary CRC and liver metastasis on CRLM patient prognosis.

Methods

In vitro study

Cell culture

The HCT116 colon cancer cell line was obtained from the Riken Cell Bank, and the hepatic stellate cell line LX2 was obtained from Cellular Engineering Technologies Inc. HCT116 cells were cultured in McCoy’s 5A Modified Medium (Life Technologies Ltd., Tokyo, Japan) with 10% fetal bovine serum (FBS) (Life Technologies Ltd.). LX2 cells were cultured in Dulbecco’s modified Eagle’s medium (DMEM) (Life Technologies Ltd.) with 10% FBS. Both cell lines were cultured under 37 °C in 5% CO2.

HSC conditioned (CM) medium preparation

HCT116 cells (3.0 × 106 cells) were cultured in McCoy medium in a 10-cm dish until cell numbers reached 3.0 × 105 cells. The cell culture media was then changed to CM from cancer cells for 24 h of culture. The medium of HCT116 (3.0 × 106 cells) changed to activated HSCs conditioned medium (HSC-CM) or DMEM (control) for 24 h culture. After that, the FBS-free medium culture was followed. After 24 h, HCT116 cells were collected for experimental analyses.

Scratch assays

Cells were plated in 6-cm dishes at 3.0 × 106 cells/dish. The medium was replaced with activated HSC-CM or DMEM (control) for 24 h. After the cells had reached confluency, a plastic pipette tip was drawn across the center of the plate to produce a scratch that was 1 mm in width. After 24 h of culture in medium with 1% FBS, a phase contrast microscope was used to examine cell movement into the wound area.

Polymerase chain reaction (PCR) analysis

RNA was extracted from samples using the RNeasy Mini Kit (Qiagen, Hilden, Germany) according to the manufacturer’s instructions. cDNA was synthesized using a reverse transcription kit (Applied Biosystems, Foster City, CA, USA). The HIF-1α TaqMan gene expression assay (Hs00153153_m1, Applied Biosystems) was used, and GAPDH (4326317E, Applied Biosystems) was selected as an internal control. The StepOnePlus Real-Time PCR System (Applied Biosystems) was used to perform qRT-PCR.

Western blotting

RIPA buffer (Thermo Fisher Scientific Inc.) containing both protease inhibitor cocktail (Sigma-Aldrich, St Louis, MO, USA) and the PhosSTOP phosphatase inhibitor cocktail (Roche, Tokyo, Japan) was used to lyse cells. Protein concentrations were measured with the BCA kit (Thermo Fisher Scientific Inc.), and equal amounts of extracted proteins were separated on 10% SDS-PAGE gels and transferred onto PVDF membranes (Bio-Rad Inc., Hercules, CA, USA). The membranes were incubated with the indicated primary antibody, followed by the appropriate HRP-conjugated secondary antibody. The bands were detected by chemiluminescence (Thermo Fisher Scientific Inc.). Primary antibody against HIF-1α (diluted 1:1000; HPA001275) was purchased from Sigma-Aldrich, and primary antibody against β-actin was obtained from Sigma Chemical (St Louis, MO, USA).

Clinicopathological study

Patient selection

Seventy-five CRLM patients who underwent an initial hepatectomy at our institute from 1994 to 2015 with available surgical specimens of primary CRCs and the matched liver metastases were enrolled in this study. This study was authorized in advance by the Institutional Review Board of the University of Tokushima Graduate School (approval ID number: 2392), and all patients provided written informed consent. The participants in this study included 47 males and 28 females with a mean age of 66.5 years, ranging from 33 to 90 years in age. The numbers of patients with synchronous and metachronous liver metastases were 32 (43%) and 43 (57%), respectively. Staging and curability was defined according to the Classification of Primary Colorectal Cancer by the Colorectal Cancer Study Group of Japan [30]. T-factor was determined by tumor number, size, and vascular infiltration. Tumor stage was determined by T-, N-, and M-factors. We defined H class as the following classification: H0 class, No metastasis to liver; H1 class, ≦ 4 lesions and ≦ 5 cm; H2 class, other than H1 and H3; H3 class, > 5 lesions and > 5cm [31]. We divided surgical procedure into minor and major hepatectomy, and major hepatectomy was defined as resection of four or more liver segments [32]. All patients had not received neoadjuvant chemotherapy and follow-up period had started after hepatectomy. The mean follow-up period was 41.3 months (range 4.4–191.3 months). We examined clinicopathological features, prognosis, molecular biological malignancy, 5-year overall survival (OS), and disease-free survival (DFS).

Immunohistochemical assessment of HIF-1α

Paraffin sections (4 μm) were cut from archival formalin-fixed paraffin-embedded tissue blocks. The samples were deparaffinized and dehydrated using a graded series of ethanol solutions. Endogenous peroxidase activity was stopped through the administration of 0.3% hydrogen peroxidase and methanol for 20 min. After rinsing in phosphate-buffered saline (PBS; Fisher Scientific, Pittsburgh, PA, USA), the tissue sections were processed in a 0.01 M citrate buffer (pH 6.0) inside a heat-resistance plastic container. The sections were irradiated in a microwave oven for 25 min and then allowed to cool at room temperature. The sections were incubated with primary mouse monoclonal antibody against HIF-1α (1:500; HPA001275, Sigma-Aldrich, MO, USA) overnight at 4 °C in a humidified chamber. The sections were incubated using Daco REALTM EnvisionTM/HRP, Rabbit/Mouse (ENV), for 45 min followed by three washes in PBS. Peroxidase labeling was developed by incubating the section in 3,3′-diaminobenzidine tetrahydrochloride (DAB) for 5 min. Nuclear counterstaining was completed using Mayer’s hematoxylin solution. Cell counts were performed using a Nikon Digital Camera DXM 1200F photomicroscope at a magnification of × 200 (× 20 objective and × 10 eyepiece). The area counted in each section was randomly selected from the representative tumor field. For each section, eight areas were assessed. The staining score for HIF-1α was determined based on staining intensity (0 negative, 1 low, 2 medium, 3 high) and staining area (0, 0%; 1, 0–25%; 2, 26–50%; 3, ≥ 51%). Scores over 4 points were defined as positive expression (Fig. 1).

Statistics

All statistical analyses were performed using statistical software (JMP software, version 11; SAS Campus Drive, Cary, NC, USA). Data are expressed as the mean ± SD. Survival curves were calculated using the Kaplan–Meier method and compared using the log-rank test. Comparisons between two groups were performed by Mann–Whitney U test. Comparisons between more than three groups were calculated using one-way ANOVA with Turkey–Kramer’s test. p < 0.05 was considered to indicate statistical significance.

Results

Tumor malignancy is enhanced in the liver by HSCs

We first examined the impact of HSC-CM on HIF-1α expression in HCT116 colorectal cancer cells. We found that HSC-CM induced both HIF-1α mRNA and protein expression in HCT116 cells (p < 0.01) (Fig. 2a, b). Moreover, HSC-CM induced migration of HCT116 cancer cells in scratch assays (p < 0.01) (Fig. 2c). Together, these results suggest the possibility that cells in the cancer microenvironment such as HSCs may alter tumor malignancy in liver metastasis.

HIF-1α expression in liver metastasis determines patient prognosis

We next examined HIF-1α expression in 75 CRLM patients with primary CRC and matched liver metastasis specimens. Positive HIF-1α expression was detected in 51 (68.0%) primary CRCs and 54 (72.0%) liver metastases. We observed the following trends of HIF-1α expression from primary CRC to liver metastasis: positive to positive expression, 37 (49.3%); positive to negative expression, 14 (18.7%); negative to positive expression, 17 (22.7%); and negative to negative expression, 7 (9.3%). Tables 1 and 2 show the patient clinicopathological factors according to HIF-1α expression in primary and metastatic sites. There were no significant relationships between HIF-1α high and low expression groups in both primary and metastatic sites. The adjuvant chemotherapy was introduced in 45 cases (60%) of all patients. According to detail regimen of the chemotherapy, we performed 5-FU-based chemotherapy in most of patients and showed the detail as follows: FOLFOX, 8 cases; LV/UFT, 8 cases; LV/5FU, 7 cases; MMC/5FU, 7 cases; IRIS, 7 cases; arterial injection. 3 cases; oral 5FU, 2 cases; and other, 3 cases.
Table 1
Clinicopathological factors according to HIF-1α expression in metastatic site
Factors
 
Low expression (n = 21)
High expression (n = 54)
p value
Primary factors
 Location
Colon/rectum
12/9
34/20
0.6433
 Depth
< SS/≥ SS
2/19
5/49
0.9718
 Diff.
Diff/undiff
20/1
51/3
0.8896
 Lymphatic invasion
−/+
9/12
20/33
0.6614
 Vessel invasion
−/+
5/16
17/37
0.5068
 LN metastasis
−/+
8/13
24/30
0.6165
Metastatic factors
 Meta. period
Meta/syn
10/11
22/32
0.5895
 Tumor size (cm)
< 5/≥ 5
15/6
42/12
0.5678
 Tumor number
< 5/≥ 5
17/4
43/11
0.8973
 H class
H1/H2.3
12/9
34/20
0.6433
 Grade
A/B.C
12/9
26/28
0.4836
 Adjuvant therapy
−/+
8/13
22/32
0.8337
 Hepatectomy
Minor/major
16/5
46/8
0.3555
 CA19-9a
< 100/≥ 100
16/4
43/4
0.2014
 CEAa
< 10/≥ 10
12/8
27/20
0.8461
Diff differentiated type, undiff undifferentiated type, LN lymph node, SS subserous, Meta metachronous, Syn synchronous
a8 patients are not available
Table 2
Clinicopathological factors according to HIF-1α expression in primary site
Factors
 
Low expression (n = 24)
High expression (n = 51)
p value
Primary factors
 Location
Colon/rectum
16/8
30/21
0.5130
 Depth
< SS/≥ SS
1/23
6/45
0.2601
 Diff.
Diff/undiff
23/1
48/3
0.7528
 Lymphatic invasion
−/+
10/14
19/31
0.6476
 Vessel invasion
−/+
6/18
16/35
0.5684
 LN metastasis
−/+
9/15
23/28
0.5334
Metastatic factors
 Meta. period
Meta/syn
7/17
25/26
0.1005
 Tumor size (cm)
< 5≥ 5
18/6
39/12
0.8896
 Tumor number
< 5/≥ 5
19/5
41/10
0.9018
 H class
H1/H2.3
14/10
32/19
0.7150
 Grade
A/B.C
11/13
27/24
0.5656
 Adjuvant therapy
−/+
8/16
22/29
0.4188
 Hepatectomy
Minor/major
17/7
45/6
0.1633
 CA19-9a
< 100/≥ 100
19/1
40/7
0.2196
 CEAa
< 10/≥ 10
10/10
29/18
0.3758
Diff differentiated type, undiff undifferentiated type, LN lymph node, SS subserous, Meta metachronous, Syn synchronous
a8 patients are not available
Regarding survival, there were no differences in OS and DFS according to HIF-1α expression in the primary site (p = 0.64 (95%CI 0.55–2.85), p = 0.91 (95%CI 0.56–1.83), respectively) (Fig. 3a, b). The percentage of patients according to HIF-1α expression (Low/High) in the primary site were 67.2/69.7% and 60.4/54.6% for 3 and 5 years in OS, and 45.8/62.2%, 33.3/31.2% for 1 and 3 years in DFS, respectively. In contrast, HIF-1α expression in the metastatic site significantly correlated with poor prognosis in both OS and DFS (p = 0.02 (95%CI 1.24–11.17), p < 0.01 (95%CI 1.30–5.16), respectively) (Fig. 4a, b). The percentage of patients according to HIF-1α expression (Low/High) in the metastatic site were 95.2/58.6% and 75.8/54.9% for 3 and 5 years in OS and 71.4/51.1% and 52.2/19.6% for 1 and 3 years in DFS. More importantly, in 41.3% of patients, HIF-1α expression was altered from primary CRC to liver metastasis, and the patients with positive HIF-1α expression in liver metastasis had significantly poor prognosis (Fig. 5a, b).
Univariate analysis of OS revealed that differentiation type (undifferentiated type, p < 0.0001) in primary tumors and H class (2.3, p = 0.0400), grade (B.C, p = 0.0119), and HIF-1α expression (positive, p = 0.0220) in metastatic tumors were significant prognostic factors (Table 3). In multivariate analysis, undifferentiated type (HR 20.873, p = 0.0013) in primary tumors and high HIF-1α expression (HR 2.850, p = 0.0422) in metastatic tumors were independent prognostic factors (Table 3).
Table 3
Prognostic factors of overall survival
Factors
5-year survival (%)
Univariate
Multivariate
HR (95% CI)
p value
HR (95% CI)
p value
Primary factors
 Location
Colon/rectum
56.0/58.0
1.03 (0.46–2.24)
0.9357
  
 Depth
< SS/> SS
65.6/58.9
0.93 (0.28–5.81)
0.9242
  
 Diff.
Diff/undiff
59.1/0
23.90 (4.58–112.20)
< 0.0001
20.87 (3.78–107.13)
0.0013
 Lymphatic invasion
−/+
66.5/53.7
0.93 (0.43–2.13)
0.9569
  
 Vessel invasion
−/+
59.9/56.7
1.16 (0.53–2.75)
0.7172
  
 LN metastasis
−/+
70.3/48.7
1.53 (0.70–3.59)
0.3019
  
 HIF-1α
−/+
60.4/54.6
1.21 (0.55–2.85)
0.6424
  
Metastatic factors
 Meta. period
Meta/syn
69.0/49.8
1.79 (0.79–4.59)
0.1826
  
 Tumor size (cm)
< 5/> 5
60.5/47.2
1.85 (0.81–4.04)
0.1213
  
 Tumor number
< 5/> 5
58.4/51.1
1.67 (0.68–3.72)
0.2231
  
 H class
H1/H2.3
63.8/46.0
2.22 (1.02–4.96)
0.0400
1.19 (0.41–4.31)
0.7591
 Grade
A/B.C
72.7/44.2
2.89 (1.27–7.42)
0.0119
2.37 (0.20-0.61)
0.1996
 Adjuvant therapy
−/+
68.3/54.4
1.22 (0.53–3.13)
0.6514
  
 Hepatectomy
Minor/major
52.6/64.7
0.68 (0.20–1.79)
0.4832
  
 HIF-1α
−/+
75.8/54.9
3.26 (1.24–11.17)
0.0220
2.85 (1.04–10.01)
0.0422
 CA19-9
< 100/> 100
28.2/37.5
0.73 (0.27–2.53)
0.2557
  
 CEA
< 10/> 10
57.4/55.1
1.45 (0.60–3.84)
0.4164
  
Diff differentiated type, undiff undifferentiated type, LN lymph node, SS subserous, Meta metachronous, Syn synchronous, HR hazard ratio, CI confidence interval
Univariate analysis of DFS revealed that differentiation type (undifferentiated type, p = 0.0466) and lymph node metastases (positive, p = 0.0146) in primary tumors and grade (B.C, p = 0.0119) and HIF-1α expression (positive, p = 0.0073) in metastatic tumors were significant recurrent factors (Table 4). In multivariate analysis, lymph node metastases (HR 2.03, p = 0.0186) in primary tumors and grade B.C (HR 2.21, p = 0.0057) and high HIF-1α expression (HR 2.40, p = 0.0079) in metastatic tumors were independent recurrent factors (Table 4).
Table 4
Prognostic factors of disease-free survival
Factors
3-year survival (%)
Univariate
Multivariate
HR (95% CI)
p value
HR (95% CI)
p value
Primary factors
 Location
Colon/rectum
27.3/30.7
1.17 (0.67–2.10)
0.4980
  
 Depth
< SS/> SS
18.8/31.2
0.77 (0.23–1.91)
0.6370
  
 Diff.
Diff/undiff
31.1/0
3.16 (1.00–8.92)
0.0466
2.14 (0.50–6.27)
0.2650
 Lymphatic invasion
−/+
32.6/31.0
1.05 (0.58–1.87)
0.7784
  
 Vessel invasion
−/+
28.0/32.7
0.92 (0.50–1.64)
0.9157
  
 LN metastasis
−/+
50.7/18.4
1.91 (1.07–3.51)
0.0146
2.03 (1.12–3.79)
0.0186
 HIF-1α
−/+
33.3/31.2
1.03 (0.56–1.83)
0.9122
  
Metastatic factors
 Meta. period
Meta/syn
39.6/23.8
1.71 (0.97–3.13)
0.0693
  
 Tumor size (cm)
< 5/> 5
26.1/41.7
0.95 (0.52–1.86)
0.8708
  
 Tumor number
< 5/> 5
30.8/27.8
1.58 (0.79–2.94)
0.1647
  
 H class
H1/H2.3
31.3/27.9
1.55 (0.88–2.70)
0.1224
  
 Grade
A/B.C
37.6/22.7
2.02 (1.16–3.58)
0.0119
2.21 (1.26–3.94)
0.0057
 Adjuvant therapy
−/+
30.2/23.8
1.54 (0.96–3.34)
0.1681
  
 Hepatectomy
Minor/major
28.8/46.2
0.97 (0.44–1.92)
0.9361
  
 HIF-1α
−/+
52.2/19.6
2.48 (1.30–5.16)
0.0073
2.40 (1.25–5.01)
0.0079
 CA19-9
< 100/> 100
63.5/37.5
1.71 (0.74–4.98)
0.5631
  
 CEA
< 10/> 10
27.6/30.1
1.19 (0.66–2.20)
0.5739
  
Diff differentiated type, undiff undifferentiated type, LN lymph node, SS subserous, Meta metachronous, Syn synchronous, HR hazard ratio, CI confidence interval

Discussion

The present study revealed that activated HSCs increased HIF-1α expression at the mRNA and protein levels and promoted tumor cell activities. The positive rate of HIF-1α expression was similar in primary CRC (68.0%) and liver metastasis (72.0%). Nevertheless, in 41.3% of patients, HIF-1α expression was altered from primary CRC to liver metastasis, and the patients with positive HIF-1α expression in liver metastasis had significantly poor prognosis. HIF-1α expression in primary CRC did not influence any malignant behavior including prognostic outcome.
Previous studies reported differences in the immune microenvironment between the primary CRC and liver metastasis, and more CD33+ cells and CD8+ cells, but not CD8+ T cells in liver metastases; these results suggested that increased numbers of immunosuppressing cells in the liver may contribute to the poor response to immunotherapy [9]. The presence of liver metastases was associated with fewer infiltrating CD8+ T cells and poor response to PD-1 therapy in other cancer types [33]. CRC-associated DNA hypomethylation undergoes hypermethylation in liver metastases [11]. Therefore, immune cells and epigenetic modifications might correlate to the alteration of positive HIF-1 expression in liver metastasis and poor prognosis of patients with positive HIF-1 expression in liver metastasis.
In the present study, undifferentiated type in primary tumors and HIF-1α high expression in metastatic tumors were the independent prognostic factors in OS. As the independent recurrent factor, lymph node metastases in primary factors and synchronous, grade B.C, and high HIF-1α expression in metastatic tumors were observed. Previous studies reported that lymph node metastases, lymphovascular invasion, and poorly differentiated type in primary tumors and the number of liver metastases were independent poor prognostic factors for progression-free survival and OS in CRLM patients [3436]. The incidence of synchronous metastasis remains high with poor survival outcomes compared with patients of metachronous metastasis [37].
Hepatocytes, Kupffer cells, and HSCs are important cells in the microenvironment of liver. In CRLM, Kupffer cells and HSCs are activated by cancer cells, and these activated stromal cells interact with metastatic cancer cells and promote tumor invasion [17]. As we previously reported that activated HSCs promote cancer cell progression through paracrine or autocrine IL-6 [18], it is known that activated HSC regulate downstream pathways and promote tumor growth [38, 39]. Our further research revealed a correlation between IL-6 expression and HIF-1α expression, but there is no significantly difference (data not shown). As our supportive opinion, other reports showed that exosomes from activated HSCs induce HIF-1α expression and affect the metabolic switch of liver nonparenchymal cells [40]. Another report showed that tamoxifen decreases the levels of HIF-1α expression by suppressing activated HSC [41]. However, as it is still unknown how activated HSC induced HIF-1α expression in cancer cell, further studies on the relationship of HSC and HIF-1α are required.
To our knowledge, this is the first study focusing on HSCs in the microenvironment and comparing HIF-1α expression between primary CRC and liver metastases. However, this study had some limitations, including its retrospective design and the small sample size. Furthermore, as we do not have the data of period from surgery to the beginning of adjuvant chemotherapy, we were not able to compare chemotherapy response with the alterations of HIF-1α expression. Our results only demonstrated a relationship between HSCs and cancer cells. Since other important cells are present in the cancer microenvironment such as TAMs and immune cells, further research on these cells is required. Our results suggest that HIF-1α expression in liver metastasis is not associated with the primary CRC and may be a useful prognostic marker. These findings should be confirmed in future studies.

Conclusions

HIF-1α expression in liver metastasis, but not primary CRC, is correlated with poor prognosis of patients with CRLM, and HSCs might play a key role in the aggressive phenotype of tumor cells. These findings may improve our understanding of various molecular alterations in metastatic tumors and guide the development of novel targeted drugs.

Acknowledgments

We thank Edanz Group (https://​en-author-services.​edanzgroup.​com/​) for editing a draft of this manuscript.
This study was approved by the Institutional Review Board of the University of Tokushima Graduate School (approval ID number: 2392). All patients have given informed consent in accordance with the ethical standards.
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.
Literatur
1.
Zurück zum Zitat Choti MA, Sitzmann JV, Tiburi MF, Sumetchotimetha W, Rangsin R, Schulick RD, et al. Trends in long-term survival following liver resection for hepatic colorectal metastases. Ann Surg. 2002;235(6):759–66.PubMedPubMedCentralCrossRef Choti MA, Sitzmann JV, Tiburi MF, Sumetchotimetha W, Rangsin R, Schulick RD, et al. Trends in long-term survival following liver resection for hepatic colorectal metastases. Ann Surg. 2002;235(6):759–66.PubMedPubMedCentralCrossRef
2.
Zurück zum Zitat Cummings LC, Payes JD, Cooper GS. Survival after hepatic resection in metastatic colorectal cancer: a population-based study. Cancer. 2007;109(4):718–26.PubMedCrossRef Cummings LC, Payes JD, Cooper GS. Survival after hepatic resection in metastatic colorectal cancer: a population-based study. Cancer. 2007;109(4):718–26.PubMedCrossRef
3.
Zurück zum Zitat Kavlakoglu B, Ustun I, Oksuz O, Pekcici R, Ergocen S, Oral S. Surgical treatment of liver metastases from colorectal cancer: experience of a single institution. Arch Iran Med. 2011;14(2):120–5.PubMed Kavlakoglu B, Ustun I, Oksuz O, Pekcici R, Ergocen S, Oral S. Surgical treatment of liver metastases from colorectal cancer: experience of a single institution. Arch Iran Med. 2011;14(2):120–5.PubMed
4.
Zurück zum Zitat Michael GH, Hiromichi I, Mithat G, Yuman F, Peter JA, Ronald PD, et al. Survival after hepatic resection for metastatic colorectal cancer: trends in outcomes for 1600 patients during two decades at a single institution. J Am Coll Surg. 2010;210(5):744–52.CrossRef Michael GH, Hiromichi I, Mithat G, Yuman F, Peter JA, Ronald PD, et al. Survival after hepatic resection for metastatic colorectal cancer: trends in outcomes for 1600 patients during two decades at a single institution. J Am Coll Surg. 2010;210(5):744–52.CrossRef
5.
Zurück zum Zitat Aloia TA, Vauthey JN, Loyer EM, Ribero D, Pawlik TM, Wei SH, et al. Solitary colorectal liver metastasis: resection determines outcome. Arch Surg. 2006;141(5):460–6.PubMedCrossRef Aloia TA, Vauthey JN, Loyer EM, Ribero D, Pawlik TM, Wei SH, et al. Solitary colorectal liver metastasis: resection determines outcome. Arch Surg. 2006;141(5):460–6.PubMedCrossRef
6.
Zurück zum Zitat Jensen NF, Smith DH, Nygård SB, Rømer MU, Nielsen KV, Brünner N. Predictive biomarkers with potential of converting conventional chemotherapy to targeted therapy in patients with metastatic colorectal cancer. Scand J Gastroenterol. 2012;47(3):340–55.PubMedCrossRef Jensen NF, Smith DH, Nygård SB, Rømer MU, Nielsen KV, Brünner N. Predictive biomarkers with potential of converting conventional chemotherapy to targeted therapy in patients with metastatic colorectal cancer. Scand J Gastroenterol. 2012;47(3):340–55.PubMedCrossRef
7.
Zurück zum Zitat Welch HG, Robertson DJ. Colorectal cancer on the decline—why screening can’t explain it all. N Engl J Med. 2016;374(17):1605–7.PubMedCrossRef Welch HG, Robertson DJ. Colorectal cancer on the decline—why screening can’t explain it all. N Engl J Med. 2016;374(17):1605–7.PubMedCrossRef
8.
Zurück zum Zitat Cremolini C, Schirripa M, Antoniotti C, Moretto R, Salvatore L, Masi G, et al. First-line chemotherapy for mCRC—a review and evidence-based algorithm. Nat Rev Clin Oncol. 2015;12(10):607–19.PubMedCrossRef Cremolini C, Schirripa M, Antoniotti C, Moretto R, Salvatore L, Masi G, et al. First-line chemotherapy for mCRC—a review and evidence-based algorithm. Nat Rev Clin Oncol. 2015;12(10):607–19.PubMedCrossRef
10.
Zurück zum Zitat Yin L, Li J, Ma D, Li D, Sun Y. Angiogenesis in primary colorectal cancer and matched metastatic tissues: biological and clinical implications for anti-angiogenic therapies. Oncol Lett. 2020;19(5):3558–66.PubMedPubMedCentral Yin L, Li J, Ma D, Li D, Sun Y. Angiogenesis in primary colorectal cancer and matched metastatic tissues: biological and clinical implications for anti-angiogenic therapies. Oncol Lett. 2020;19(5):3558–66.PubMedPubMedCentral
11.
Zurück zum Zitat Orjuela S, Menigatti M, Schraml P, Kambakamba P, Robinson MD, Marra G. The DNA hypermethylation phenotype of colorectal cancer liver metastases resembles that of the primary colorectal cancers. BMC Cancer. 2020;20(1):290.PubMedPubMedCentralCrossRef Orjuela S, Menigatti M, Schraml P, Kambakamba P, Robinson MD, Marra G. The DNA hypermethylation phenotype of colorectal cancer liver metastases resembles that of the primary colorectal cancers. BMC Cancer. 2020;20(1):290.PubMedPubMedCentralCrossRef
12.
Zurück zum Zitat Wu JB, Sarmiento AL, Fiset PO, Lazaris A, Metrakos P, Petrillo S, et al. Histologic features and genomic alterations of primary colorectal adenocarcinoma predict growth patterns of liver metastasis. World J Gastroenterol. 2019;25(26):3408–25.PubMedPubMedCentralCrossRef Wu JB, Sarmiento AL, Fiset PO, Lazaris A, Metrakos P, Petrillo S, et al. Histologic features and genomic alterations of primary colorectal adenocarcinoma predict growth patterns of liver metastasis. World J Gastroenterol. 2019;25(26):3408–25.PubMedPubMedCentralCrossRef
13.
Zurück zum Zitat Kim EK, Song MJ, Jung Y, Lee WS, Jang HH. Proteomic analysis of primary colon cancer and synchronous solitary liver metastasis. Cancer Genomics Proteomics. 2019;16(6):583–92.PubMedPubMedCentralCrossRef Kim EK, Song MJ, Jung Y, Lee WS, Jang HH. Proteomic analysis of primary colon cancer and synchronous solitary liver metastasis. Cancer Genomics Proteomics. 2019;16(6):583–92.PubMedPubMedCentralCrossRef
16.
Zurück zum Zitat Affo S, Yu LX, Schwabe RF. The role of cancer-associated fibroblasts and fibrosis in liver cancer. Annu Rev Pathol. 2017;12:153–86.PubMedCrossRef Affo S, Yu LX, Schwabe RF. The role of cancer-associated fibroblasts and fibrosis in liver cancer. Annu Rev Pathol. 2017;12:153–86.PubMedCrossRef
17.
Zurück zum Zitat Pnina B. Role of the microenvironment in liver metastasis: from pre-to prometastatic niche. Clin Cancer Res. 2016;22(24):5971–82.CrossRef Pnina B. Role of the microenvironment in liver metastasis: from pre-to prometastatic niche. Clin Cancer Res. 2016;22(24):5971–82.CrossRef
18.
Zurück zum Zitat Iwahasi S, Rui F, Morine Y, Yamada S, Saito YU, Ikemoto T, et al. Hepatic stellate cells contribute to the tumor malignancy of hepatocellular carcinoma through the IL-6 pathway. Anticancer Res. 2020;40(2):743–9.PubMedCrossRef Iwahasi S, Rui F, Morine Y, Yamada S, Saito YU, Ikemoto T, et al. Hepatic stellate cells contribute to the tumor malignancy of hepatocellular carcinoma through the IL-6 pathway. Anticancer Res. 2020;40(2):743–9.PubMedCrossRef
19.
Zurück zum Zitat Zhong H, De Marzo AM, Laughner E, Lim M, Hilton DA, Zagzag D, et al. Overexpression of hypoxia-inducible factor 1alpha in common human cancers and their metastases. Cancer Res. 1999;59(22):5830–5.PubMed Zhong H, De Marzo AM, Laughner E, Lim M, Hilton DA, Zagzag D, et al. Overexpression of hypoxia-inducible factor 1alpha in common human cancers and their metastases. Cancer Res. 1999;59(22):5830–5.PubMed
20.
Zurück zum Zitat Birner P, Schindl M, Obermair A, Breitenecker G, Oberhuber G. Expression of hypoxia-inducible factor 1alpha in epithelial ovarian tumors: its impact on prognosis and on response to chemotherapy. Clin Cancer Res. 2001;7(6):1661–8.PubMed Birner P, Schindl M, Obermair A, Breitenecker G, Oberhuber G. Expression of hypoxia-inducible factor 1alpha in epithelial ovarian tumors: its impact on prognosis and on response to chemotherapy. Clin Cancer Res. 2001;7(6):1661–8.PubMed
21.
Zurück zum Zitat Sumiyoshi Y, Kakeji Y, Egashira A, Mizokami K, Orita H, Maehara Y. Overexpression of hypoxia-inducible factor 1alpha and p53 is a marker for an unfavorable prognosis in gastric cancer. Clin Cancer Res. 2006;12(17):5112–7.PubMedCrossRef Sumiyoshi Y, Kakeji Y, Egashira A, Mizokami K, Orita H, Maehara Y. Overexpression of hypoxia-inducible factor 1alpha and p53 is a marker for an unfavorable prognosis in gastric cancer. Clin Cancer Res. 2006;12(17):5112–7.PubMedCrossRef
22.
Zurück zum Zitat Batmunkh E, Shimada M, Morine Y, Imura S, Kanemura H, Arakawa Y, et al. Expression of hypoxia-inducible factor-1 alpha (HIF-1alpha) in patients with the gallbladder carcinoma. Int J Clin Oncol. 2010;15(1):59–64.PubMedCrossRef Batmunkh E, Shimada M, Morine Y, Imura S, Kanemura H, Arakawa Y, et al. Expression of hypoxia-inducible factor-1 alpha (HIF-1alpha) in patients with the gallbladder carcinoma. Int J Clin Oncol. 2010;15(1):59–64.PubMedCrossRef
23.
Zurück zum Zitat Miyake K, Yoshizumi T, Imura S, Sugimoto K, Batmunkh E, Kanemura H, et al. Expression of hypoxia-inducible factor-1alpha, histone deacetylase 1, and metastasis-associated protein 1 in pancreatic carcinoma: correlation with poor prognosis with possible regulation. Pancreas. 2008;36(3):e1–9.PubMedCrossRef Miyake K, Yoshizumi T, Imura S, Sugimoto K, Batmunkh E, Kanemura H, et al. Expression of hypoxia-inducible factor-1alpha, histone deacetylase 1, and metastasis-associated protein 1 in pancreatic carcinoma: correlation with poor prognosis with possible regulation. Pancreas. 2008;36(3):e1–9.PubMedCrossRef
24.
Zurück zum Zitat Griffiths EA, Pritchard SA, Valentine HR, Whitchelo N, Bishop PW, Ebert MP, et al. Hypoxia-inducible factor-1a expression in the gastric carcinogenesis sequence and its prognostic role in gastric and gastro-esophageal adenocarcinomas. Br J Cancer. 2007;96(1):95–103.PubMedCrossRef Griffiths EA, Pritchard SA, Valentine HR, Whitchelo N, Bishop PW, Ebert MP, et al. Hypoxia-inducible factor-1a expression in the gastric carcinogenesis sequence and its prognostic role in gastric and gastro-esophageal adenocarcinomas. Br J Cancer. 2007;96(1):95–103.PubMedCrossRef
25.
Zurück zum Zitat Kim Y, Nam HJ, Lee J, Park DY, Kim C, Yu YS, et al. Methylation-dependent regulation of HIF-1a stability restricts retinal and tumor angiogenesis. Nat Commun. 2016;7:10347.PubMedPubMedCentralCrossRef Kim Y, Nam HJ, Lee J, Park DY, Kim C, Yu YS, et al. Methylation-dependent regulation of HIF-1a stability restricts retinal and tumor angiogenesis. Nat Commun. 2016;7:10347.PubMedPubMedCentralCrossRef
26.
Zurück zum Zitat Pouysségur J, Dayan F, Mazure NM. Hypoxia signaling in cancer and approaches to enforce tumor regression. Nature. 2006;441(7092):437–43.PubMedCrossRef Pouysségur J, Dayan F, Mazure NM. Hypoxia signaling in cancer and approaches to enforce tumor regression. Nature. 2006;441(7092):437–43.PubMedCrossRef
27.
Zurück zum Zitat Huihui L, Matjaz R, Longchang J, David H, Heiko H. Antagonistic effects of p53 and HIF1A on microRNA-34a regulation of PPP1R11 and STAT3 and hypoxia-induced epithelial to mesenchymal transition in colorectal cancer cells. Gastroenterology. 2017;153(2):505–20.CrossRef Huihui L, Matjaz R, Longchang J, David H, Heiko H. Antagonistic effects of p53 and HIF1A on microRNA-34a regulation of PPP1R11 and STAT3 and hypoxia-induced epithelial to mesenchymal transition in colorectal cancer cells. Gastroenterology. 2017;153(2):505–20.CrossRef
28.
Zurück zum Zitat Dupuy F, Tabariès S, Andrzejewski S, Dong Z, Blagih J, Annis MG, et al. PDK1-dependent metabolic reprogramming dictates metastatic potential in breast cancer. Cell Metabolism. 2015;22(4):577–89.PubMedCrossRef Dupuy F, Tabariès S, Andrzejewski S, Dong Z, Blagih J, Annis MG, et al. PDK1-dependent metabolic reprogramming dictates metastatic potential in breast cancer. Cell Metabolism. 2015;22(4):577–89.PubMedCrossRef
29.
Zurück zum Zitat Shimomura M, Hinoi T, Kuroda S, Adachi T, Kawaguchi Y, Sasada T, et al. Overexpression of hypoxia inducible factor-1 alpha is an independent risk factor for recurrence after curative resection of colorectal liver metastasis. Ann Sug Oncol. 2013;20(3):527–36.CrossRef Shimomura M, Hinoi T, Kuroda S, Adachi T, Kawaguchi Y, Sasada T, et al. Overexpression of hypoxia inducible factor-1 alpha is an independent risk factor for recurrence after curative resection of colorectal liver metastasis. Ann Sug Oncol. 2013;20(3):527–36.CrossRef
30.
Zurück zum Zitat Hashiguchi Y, Muro K, Saito Y, Ito Y, Ajioka Y, Hamaguchi T, et al. Japanese Society for Cancer of the Colon and Rectum (JSCCR) guidelines 2019 for the treatment of colorectal cancer. Int J Clin Oncol. 2020;25(1):1–42.PubMedCrossRef Hashiguchi Y, Muro K, Saito Y, Ito Y, Ajioka Y, Hamaguchi T, et al. Japanese Society for Cancer of the Colon and Rectum (JSCCR) guidelines 2019 for the treatment of colorectal cancer. Int J Clin Oncol. 2020;25(1):1–42.PubMedCrossRef
31.
Zurück zum Zitat Yamaguchi T, Mori T, Takahashi K, Matsumoto H, Miyamoto H, Kato T. A new classification system for liver metastases from colorectal cancer in Japanese multicenter analysis. Hepatogastroenterology. 2008;55(81):173–8.PubMed Yamaguchi T, Mori T, Takahashi K, Matsumoto H, Miyamoto H, Kato T. A new classification system for liver metastases from colorectal cancer in Japanese multicenter analysis. Hepatogastroenterology. 2008;55(81):173–8.PubMed
32.
Zurück zum Zitat Reddy SK, Barbas AS, Turley RS, Steel JL, Tsung A, Marsh JW, et al. A standard definition of major hepatectomy: resection of four or more liver segments. HPB (Oxford). 2011;13(7):494–502.PubMedPubMedCentralCrossRef Reddy SK, Barbas AS, Turley RS, Steel JL, Tsung A, Marsh JW, et al. A standard definition of major hepatectomy: resection of four or more liver segments. HPB (Oxford). 2011;13(7):494–502.PubMedPubMedCentralCrossRef
33.
Zurück zum Zitat Tumeh PC, Hellmann MD, Hamid O, Tsai KK, Loo KL, Gubens MA, et al. Liver metastasis and treatment outcome with anti-pd-1 monoclonal antibody in patients with melanoma and NSCLC. Cancer Immunol Res. 2017;5(5):417–24.PubMedPubMedCentralCrossRef Tumeh PC, Hellmann MD, Hamid O, Tsai KK, Loo KL, Gubens MA, et al. Liver metastasis and treatment outcome with anti-pd-1 monoclonal antibody in patients with melanoma and NSCLC. Cancer Immunol Res. 2017;5(5):417–24.PubMedPubMedCentralCrossRef
34.
Zurück zum Zitat Li Destri G, Barchitta M, Pesce A, Latteri S, Bosco D, Di Cataldo A, et al. Predictive value of the number of harvested lymph nodes and cut-off for lymph node ratio in the prognosis of stage II and III colorectal cancer patients. J Invest Surg. 2019;32(1):1–7.PubMedCrossRef Li Destri G, Barchitta M, Pesce A, Latteri S, Bosco D, Di Cataldo A, et al. Predictive value of the number of harvested lymph nodes and cut-off for lymph node ratio in the prognosis of stage II and III colorectal cancer patients. J Invest Surg. 2019;32(1):1–7.PubMedCrossRef
35.
Zurück zum Zitat Yonemura K, Kajiwara Y, Ao T, Mochizuki S, Shinto E, Okamoto K, et al. Prognostic value of poorly differentiated clusters in liver metastatic lesions of colorectal carcinoma. Am J Surg Pathol. 2019;43(10):1341–8.PubMedCrossRef Yonemura K, Kajiwara Y, Ao T, Mochizuki S, Shinto E, Okamoto K, et al. Prognostic value of poorly differentiated clusters in liver metastatic lesions of colorectal carcinoma. Am J Surg Pathol. 2019;43(10):1341–8.PubMedCrossRef
36.
Zurück zum Zitat Jang KU, Kim CW, Kim KH, Lim SB, Yu CS, Kim W, et al. Prognostic factors in terms of the number of metastatic nodules in patients with colorectal cancer liver metastases. Ann Coloproctol. 2016;32(3):92–100.PubMedPubMedCentralCrossRef Jang KU, Kim CW, Kim KH, Lim SB, Yu CS, Kim W, et al. Prognostic factors in terms of the number of metastatic nodules in patients with colorectal cancer liver metastases. Ann Coloproctol. 2016;32(3):92–100.PubMedPubMedCentralCrossRef
37.
Zurück zum Zitat Okholm C, Mollerup TK, Schultz NA, Strandby RB, Achiam MP. Synchronous and metachronous liver metastases in patients with colorectal cancer. Dan Med J. 2018;65(12):A5524.PubMed Okholm C, Mollerup TK, Schultz NA, Strandby RB, Achiam MP. Synchronous and metachronous liver metastases in patients with colorectal cancer. Dan Med J. 2018;65(12):A5524.PubMed
39.
Zurück zum Zitat Yin C, Evason KJ, Asahina K, Stainier DY. Hepatic stellate cells in liver development, regeneration, and cancer. J Clin Invest. 2013;123(5):1902–10.PubMedPubMedCentralCrossRef Yin C, Evason KJ, Asahina K, Stainier DY. Hepatic stellate cells in liver development, regeneration, and cancer. J Clin Invest. 2013;123(5):1902–10.PubMedPubMedCentralCrossRef
40.
Zurück zum Zitat Wan L, Xia T, Du Y, Liu J, Xie Y, Zhang Y, et al. Exosomes from activated hepatic stellate cells contain GLUT1 and PKM2: a role for exosomes in metabolic switch of liver nonparenchymal cells. FASEB J. 2019;33(7):8530–42.PubMedCrossRef Wan L, Xia T, Du Y, Liu J, Xie Y, Zhang Y, et al. Exosomes from activated hepatic stellate cells contain GLUT1 and PKM2: a role for exosomes in metabolic switch of liver nonparenchymal cells. FASEB J. 2019;33(7):8530–42.PubMedCrossRef
41.
Zurück zum Zitat Cortes E, Lachowski D, Rice A, Thorpe SD, Robinson B, Yeldag G, et al. Tamoxifen mechanically deactivates hepatic stellate cells via the G protein-coupled estrogen receptor. Oncogene. 2019;38(16):2910–22.PubMedCrossRef Cortes E, Lachowski D, Rice A, Thorpe SD, Robinson B, Yeldag G, et al. Tamoxifen mechanically deactivates hepatic stellate cells via the G protein-coupled estrogen receptor. Oncogene. 2019;38(16):2910–22.PubMedCrossRef
Metadaten
Titel
HIF-1α expression in liver metastasis but not primary colorectal cancer is associated with prognosis of patients with colorectal liver metastasis
verfasst von
Yuma Wada
Yuji Morine
Satoru Imura
Tetsuya Ikemoto
Yu Saito
Chie Takasu
Shinichiro Yamada
Mitsuo Shimada
Publikationsdatum
01.12.2020
Verlag
BioMed Central
Erschienen in
World Journal of Surgical Oncology / Ausgabe 1/2020
Elektronische ISSN: 1477-7819
DOI
https://doi.org/10.1186/s12957-020-02012-5

Weitere Artikel der Ausgabe 1/2020

World Journal of Surgical Oncology 1/2020 Zur Ausgabe

Vorsicht, erhöhte Blutungsgefahr nach PCI!

10.05.2024 Koronare Herzerkrankung Nachrichten

Nach PCI besteht ein erhöhtes Blutungsrisiko, wenn die Behandelten eine verminderte linksventrikuläre Ejektionsfraktion aufweisen. Das Risiko ist umso höher, je stärker die Pumpfunktion eingeschränkt ist.

Darf man die Behandlung eines Neonazis ablehnen?

08.05.2024 Gesellschaft Nachrichten

In einer Leseranfrage in der Zeitschrift Journal of the American Academy of Dermatology möchte ein anonymer Dermatologe bzw. eine anonyme Dermatologin wissen, ob er oder sie einen Patienten behandeln muss, der eine rassistische Tätowierung trägt.

Deutlich weniger Infektionen: Wundprotektoren schützen!

08.05.2024 Postoperative Wundinfektion Nachrichten

Der Einsatz von Wundprotektoren bei offenen Eingriffen am unteren Gastrointestinaltrakt schützt vor Infektionen im Op.-Gebiet – und dient darüber hinaus der besseren Sicht. Das bestätigt mit großer Robustheit eine randomisierte Studie im Fachblatt JAMA Surgery.

Chirurginnen und Chirurgen sind stark suizidgefährdet

07.05.2024 Suizid Nachrichten

Der belastende Arbeitsalltag wirkt sich negativ auf die psychische Gesundheit der Angehörigen ärztlicher Berufsgruppen aus. Chirurginnen und Chirurgen bilden da keine Ausnahme, im Gegenteil.

Update Chirurgie

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

S3-Leitlinie „Diagnostik und Therapie des Karpaltunnelsyndroms“

Karpaltunnelsyndrom BDC Leitlinien Webinare
CME: 2 Punkte

Das Karpaltunnelsyndrom ist die häufigste Kompressionsneuropathie peripherer Nerven. Obwohl die Anamnese mit dem nächtlichen Einschlafen der Hand (Brachialgia parästhetica nocturna) sehr typisch ist, ist eine klinisch-neurologische Untersuchung und Elektroneurografie in manchen Fällen auch eine Neurosonografie erforderlich. Im Anfangsstadium sind konservative Maßnahmen (Handgelenksschiene, Ergotherapie) empfehlenswert. Bei nicht Ansprechen der konservativen Therapie oder Auftreten von neurologischen Ausfällen ist eine Dekompression des N. medianus am Karpaltunnel indiziert.

Prof. Dr. med. Gregor Antoniadis
Berufsverband der Deutschen Chirurgie e.V.

S2e-Leitlinie „Distale Radiusfraktur“

Radiusfraktur BDC Leitlinien Webinare
CME: 2 Punkte

Das Webinar beschäftigt sich mit Fragen und Antworten zu Diagnostik und Klassifikation sowie Möglichkeiten des Ausschlusses von Zusatzverletzungen. Die Referenten erläutern, welche Frakturen konservativ behandelt werden können und wie. Das Webinar beantwortet die Frage nach aktuellen operativen Therapiekonzepten: Welcher Zugang, welches Osteosynthesematerial? Auf was muss bei der Nachbehandlung der distalen Radiusfraktur geachtet werden?

PD Dr. med. Oliver Pieske
Dr. med. Benjamin Meyknecht
Berufsverband der Deutschen Chirurgie e.V.

S1-Leitlinie „Empfehlungen zur Therapie der akuten Appendizitis bei Erwachsenen“

Appendizitis BDC Leitlinien Webinare
CME: 2 Punkte

Inhalte des Webinars zur S1-Leitlinie „Empfehlungen zur Therapie der akuten Appendizitis bei Erwachsenen“ sind die Darstellung des Projektes und des Erstellungswegs zur S1-Leitlinie, die Erläuterung der klinischen Relevanz der Klassifikation EAES 2015, die wissenschaftliche Begründung der wichtigsten Empfehlungen und die Darstellung stadiengerechter Therapieoptionen.

Dr. med. Mihailo Andric
Berufsverband der Deutschen Chirurgie e.V.