Skip to main content
Erschienen in: BMC Cancer 1/2023

Open Access 01.12.2023 | Research

High expression of PPFIA1 in human esophageal squamous cell carcinoma correlates with tumor metastasis and poor prognosis

verfasst von: Yongyin Gao, Lizhao Guan, Ran Jia, Wanyi Xiao, Youming Han, Yue Li, Peng Tang, Zhentao Yu, Hongdian Zhang

Erschienen in: BMC Cancer | Ausgabe 1/2023

Abstract

Background

PTPRF interacting protein alpha 1 (PPFIA1) is reportedly related to the occurrence and progression of several kinds of malignancies. However, its role in esophageal squamous cell carcinoma (ESCC) is unclear. This current study investigated the prognostic significance and biological functions of PPFIA1 in ESCC.

Methods

Oncomine, Gene Expression Profiling Interactive Analysis (GEPIA), and Gene Expression Omnibus (GEO) were used to investigate PPFIA1 expression in esophageal cancer. The relationship between PPFIA1 expression and clinicopathological characteristics and patient survival was evaluated in GSE53625 dataset, and verified in the cDNA array based on qRT–PCR and tissue microarray (TMA) dataset based on immunohistochemistry. The impact of PPFIA1 on the migration and invasion of cancer cells were investigated by wound-healing and transwell assays, respectively.

Results

The expression of PPFIA1 was obviously increased in ESCC tissues versus adjacent esophageal tissues according to online database analyses (all P < 0.05). High PPFIA1 expression was closely related to several clinicopathological characteristics, including tumor location, histological grade, tumor invasion depth, lymph node metastasis, and tumor-node-metastasis (TNM) stage. High PPFIA1 expression was related to worse outcomes and was identified as an independent prognostic factor of overall survival in ESCC patients (GSE53625 dataset, P = 0.019; cDNA array dataset, P < 0.001; TMA dataset, P = 0.039). Downregulation of PPFIA1 expression can significantly reduce the migration and invasion ability of ESCC cells.

Conclusion

PPFIA1 is related to the migration and invasion of ESCC cells, and can be used as a potential biomarker to evaluate the prognosis of ESCC patients.
Hinweise

Supplementary Information

The online version contains supplementary material available at https://​doi.​org/​10.​1186/​s12885-023-10872-9.
Yongyin Gao and Lizhao Guan contributed equally to this work and considered co-first authors.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Abkürzungen
ESCC
Esophageal squamous cell carcinoma
PPFIA1
PTPRF interacting protein alpha 1
GEPIA
Gene Expression Profiling Interactive Analysis
TMA
Tissue microarray
PPI
Protein–protein interaction
STRING
Search Tool for the Retrieval of Interacting Genes
RFS
Recurrence-free survival
PFS
Progression-free survival
FP
Time to first progression
OS
Overall survival
PPS
Post-progression survival
DMFS
Distant metastasis-free survival

Background

Esophageal cancer is ranked as the eighth most common cancer and the sixth leading cause of cancer-related death worldwide [1]. Esophageal squamous cell carcinoma (ESCC) is the main pathological subtype, accounting for nearly 90% of esophageal cancer tumors in China [2]. Surgical resection and neoadjuvant or adjuvant therapy are the main methods for ESCC treatment. Despite advances in diagnosis and therapy, recurrence and metastasis are common and significantly decrease the overall survival (OS) of patients [3]. Therefore, there is an urgent need to identify the metastatic mechanisms of ESCC and to explore effective indicators for early diagnosis, prognosis prediction, and treatment response evaluation.
PTPRF interacting protein alpha 1 (PPFIA1) is a member of the LAR protein-tyrosine phosphatase-interacting protein family of cytosolic scaffold proteins [4]. Several studies have reported that PPFIA1 is abnormally expressed, plays a vital role in invasion and metastasis, and may be used as a potential prognostic molecular marker for several types of malignancies, including breast cancer, laryngeal cancer, oral squamous cell carcinoma, colon cancer and ovarian cancer [510]. However, the expression of PPFIA1 and its correlation with clinicopathological features and patient prognosis in ESCC remain uncertain.
In the present research, we first investigated the expression of PPFIA1 in ESCC tissues and paracancerous tissues using available online datasets from the Oncomine, Gene Expression Profiling Interactive Analysis (GEPIA), and Gene Expression Omnibus (GEO) databases. The associations of PPFIA1 expression with the clinicopathological features and outcomes of ESCC patients were then analyzed in the GSE53625 dataset and further confirmed in cDNA array and tissue microarray (TMA) datasets. Finally, the effects of PPFIA1 on the migration and invasion of ESCC cells were investigated.

Materials and methods

GEPIA

The expression of PPFIA1 mRNA in human tumors was analyzed through the GEPIA website (http://​gepia.​cancer-pku.​cn/​detail.​php), which contains RNA sequencing and expression data from The Cancer Genome Atlas (TCGA) and the Genotype-Tissue Expression (GTEx) databases [11].

Oncomine

Oncomine (http://​www.​oncomine.​org) is a public online microarray database and comprehensive data mining platform that can be used to mine cancer genetic information [12].

GEO

The GEO database (https://​www.​ncbi.​nlm.​nih.​gov/​geo/​) is a publicly accessible database of gene expression data that stores a large amount of microarray data [13]. The GEO datasets were analyzed online through GEO2R.
Furthermore, the RNA sequencing data, all clinicopathological variable data and the survival data of 179 ESCC patients were also downloaded from the GSE53625 dataset (https://​www.​ncbi.​nlm.​nih.​gov/​geo/​query/​acc.​cgi?​acc=​GSE53625). There were 146 (81.6%) males and 33 (18.4%) females, with a median age of 60 (range, 36 ~ 82) years. According to the criteria of the seventh edition of the American Joint Committee on Cancer (AJCC) tumor-node-metastasis (TNM) staging system for ESCC, 10 (5.6%), 77 (43.0%), and 92 (51.4%) cases were considered stage I, II, and III, respectively.

TCGA

The RNA-sequencing data and clinical information of 81 ESCC patients and 11 normal esophageal samples were downloaded from TCGA database (https://​cancergenome.​nih.​gov/​). The level of gene expression was measured as fragments per kilobase of transcript per million mapped reads (FPKM) [14].

Kaplan–Meier analysis

Kaplan–Meier Plotter (www.​kmplot.​com) is an online database containing survival information (including recurrence-free survival [RFS], progression-free survival [PFS], time to first progression [FP], OS, postprogression survival [PPS], and distant metastasis-free survival [DMFS]) for patients with breast, ovarian, lung or gastric cancers. The cancer samples were divided into two groups (low vs. high PPFIA1 expression) based on the automatically selected best cutoff values.

Search Tool for the Retrieval of Interacting Genes (STRING)

We further used the STRING website (http://​string-db.​org/​) to explore the interactions between PPFIA1 and other genes based on a generated protein–protein interaction (PPI) network [15].

Analysis of tissue cDNA array data based on quantitative real-time polymerase chain reaction (qRT-PCR)

An ESCC cDNA microarray with patient diagnosis information was purchased from Shanghai Outdo Biotech Co., Ltd. (Cat. No.: cDNA-HEsoS095Su01; Shanghai, China); the array contained samples from 67 cancer tissues and 28 adjacent esophageal tissues. These tissues were all from patients who were histologically confirmed to have ESCC and underwent esophagectomy. The tissues were from 48 males and 19 females, with a median age of 59 (range, 37 ~ 78) years. The study was approved by the Medical Ethics Committee of the Shanghai Outdo Biotech Company.
Total RNA was isolated using TRIzol reagent (Takara, Dalian, China) and the cDNA was then synthesized using the SuperScript™ II Reverse Transcriptase kit (Thermo Fisher Scientific, Waltham, MA, USA) according to the manufacturer’s instructions. Then, 2 µl cDNA was used as a template for qRT–PCR with SYBR® Premix Ex Taq™ II (Takara, Dalian, China), and β-actin was used as the internal control. The primer sequences used were as follows: PPFIA1, forward 5'-CTTAACCCAGGGGAAGTTACAC-3' and reverse 5'-ATCCTAAGAGACCGCTCATGC-3'; and β-actin, forward 5'-GAAGAGCTACGAGCTGCCTGA-3' and reverse 5'-CAGACAGCACTGTGTTGGCG-3'. The expression fold-changes were quantified by the 2−∆∆CT method.

Immunohistochemistry (IHC) analysis

TMAs containing samples from 147 ESCC tissues and 40 adjacent normal esophageal tissues were made by Shanghai Outdo Biotech Co. Ltd. All paraffin block specimens were obtained from patients who underwent radical esophagectomy between 2009 and 2010 at Tianjin Medical University Cancer Institute and Hospital, with reliable information on survival. No patients had received any chemotherapy or radiotherapy before surgery. Among the patients, there were 119 males and 28 females with a median age of 68 years old. All patients were followed up until September 2016 with a median follow-up period of 36 months. The research protocol was approved by the Research Ethics Committee of Tianjin Medical University Cancer Institute and Hospital, and written informed consent was obtained from all patients.
The protein expression of PPFIA1 was detected through IHC. In brief, the tissue sections were deparaffinized, rehydrated and incubated with an antigen retrieval solution. The activity of endogenous peroxidase was blocked with 0.3% H2O2 and 5% goat serum. Then, the sections were incubated with an anti-PPFIA1 polyclonal antibody (1:100; Cat No. DF12102) at 4 °C overnight and then incubated with a biotinylated secondary antibody for 20 min at room temperature. Diaminobenzidine was used as a chromogen. The stained slide was scanned using an automatic slice scanning system.
Two or three experienced pathologists who were blinded to the clinicopathological information independently performed the staining evaluation based on the staining intensity (no staining, 0; weak, 1; moderate, 2; strong, 3) and the percentage (0%, 0; 1% ~ 25% positive, 1; 26% ~ 50% positive, 2; 51% ~ 75% positive, 3; 76% ~ 100% positive, 4) of positively stained tumor cells. The immunoreactivity score was calculated as the product of the staining intensity and staining percentage scores, with the final score ranging from 0 to 12. Patients with a total score of < 4 were considered to have low expression, and those with a total score of ≥ 4 were considered to have high expression.

Cell culture and transfection

The human ESCC cell lines Kyse-30 and Ec-109 were grown in RPMI 1640 medium containing 10% fetal bovine serum (Gibco, USA). The siRNA targeting PPFIA1 (si-PPFIA1) and negative control were synthesized by GenePharma (Shanghai, China). ESCC cells were transiently transfected with these siRNAs using Lipofectamine 2000 reagent (Invitrogen, Life Technologies) following the manufacturer's instructions. 48 h after transfection, the RNA and protein were harvested and analyzed.

Western blotting

Western blotting was performed as described previously [16]. Total protein was extracted using RIPA buffer supplemented with protease inhibitors. The protein concentration was detected by a BCA Reagent Kit. Equal amount of protein was electrophoresed and transferred onto PVDF membranes. Non-specific binding was blocked with 5% skim milk. The membranes were then incubated with primary antibodies overnight at 4 °C and secondary antibodies at room temperature for 2 h. Protein expression was visualized with an enhanced chemiluminescence system.

Cell migration and invasion assays

The wound-healing assay was conducted to evaluate cellular migration according to our published protocol [17]. ESCC cells were seeded into 6-well plates. When the cell confluence achieved about 80 ~ 90%, scratches were created by scraping the cell layer using a 10 μl pipette tips across each well. After washing with PBS, the cells were cultured with serum-free medium at 37 °C containing 5% CO2. The remaining cells were cultured for another 48 h.
Cells which suspended with serum-free medium were plated into the upper chamber of an insert Transwell chambers (Corning, NY, USA) with or without coated Matrigel (BD Biosciences, CA, USA). The lower chambers were filled with medium containing 10% FBS. After incubation for 36 h at 37 °C, migrated or invasive cells on the lower surface of the membrane were fixed and stained with crystal violet, and cells in five randomly selected fields were photographed and counted.

Statistical analyses

Statistical analyses were performed using SPSS 17.0 software (SPSS, Inc., Chicago, IL) and GraphPad Prism 7.0 software (GraphPad, Inc., La Jolla, CA, USA). Data were expressed as the mean ± standard deviation (SD). The differences between the groups were assessed using Student's t test. The patients were divided into two groups according to the best cutoff value of PPFIA1 expression for predicting OS calculated with the X-tile 3.6.1 software (Yale University, New Haven, CT, USA) for analysis of the GSE53625 dataset [18] or according to the median value of PPFIA1 expression for analysis of the cDNA array dataset. The chi-square test or Fisher’s exact test was used for the analysis of PPFIA1 expression and clinicopathological variables. Survival analysis was carried out through the Kaplan–Meier method and the log-rank test. The correlations between PPFIA1 expression and the expression of other genes were assessed by Pearson’s correlation analysis. A P value < 0.05 was deemed statistically significant.

Results

The expression of PPFIA1 mRNA in various tumor tissues

We first examined the expression level of PPFIA1 mRNA in various tumors with the GEPIA website, and the results showed significantly higher PPFIA1 mRNA expression in several types of tumor tissues, including esophageal cancer (ESCA), pancreatic adenocarcinoma (PAAD), and thymoma tissues, compared with adjacent normal tissues (Fig. 1A). Overall PPFIA1 expression in various tumor specimens, as well as in normal controls, were further analyzed with the Oncomine database. PPFIA1 mRNA expression was obviously increased in the tumor tissues versus normal tissues from most of the datasets for all types of tumors (Fig. 1B). The fold changes in PPFIA1 mRNA expression in different ESCA tissues of 7 datasets are displayed in Supplementary Table 1.

The expression of PPFIA1 was upregulated in ESCC tissues

The expression of PPFIA1 mRNA was markedly higher in ESCA tumor samples than in adjacent normal samples according to the GEPIA website (Fig. 2A). This finding was further verified using data from six GEO datasets; the results indicated that the relative PPFIA1 mRNA expression was obviously upregulated in ESCC tissues compared with paired or unpaired adjacent normal tissues in the GSE23400 (9.6 ± 1.1 vs. 8.4 ± 0.6, P < 0.001, Fig. 2B), GSE20347 (10.3 ± 1.3 vs. 8.4 ± 0.2, P < 0.001, Fig. 2C), GSE29001 (6395.6 ± 4748.9 vs. 1498.6 ± 337.9, P = 0.005, Fig. 2D), GSE53625 (12.6 ± 1.2 vs. 11.0 ± 0.5, P < 0.001, Fig. 2E), GSE45670 (251.5 ± 185.9 vs. 97.5 ± 49.6, P = 0.015, Fig. 2F) and GSE26886 (1.3 ± 1.4 vs. 0.1 ± 1.1, P = 0.022, Fig. 2G) datasets.
In addition, the increased expression of PPFIA1 mRNA in ESCC samples was further confirmed through the analysis of cDNA array data based on qRT–PCR (P = 0.003, Fig. 2H).

Correlations between PPFIA1 mRNA expression and clinicopathological variables in ESCC

In the GSE53625 dataset, the 179 ESCC patients were divided into a low-expression group (n = 155) and a high-expression group (n = 24) according to the X-tile cutoff. Our results showed that high PPFIA1 expression was positively related to tumor invasion depth (P = 0.019), lymph node metastasis (P = 0.013), and TNM stage (P = 0.012), but no correlations were found between PPFIA1 expression and gender, age, smoking use, alcohol use, tumor location, histological grade or adjuvant therapy (P > 0.05) (Table 1).
Table 1
Associations of PPFIA1 mRNA expression with clinicopathological variables of 179 ESCC patients from the GSE53625 dataset
Clinicopathological variables
Number
PPFIA1 expression
x2
P value
Low
High
Gender
   
0.649
0.420
 Male
146
125
21
  
 Female
33
30
3
  
Age (years)
   
0.316
0.574
 ≤ 60
99
87
12
  
 ˃60
80
68
12
  
Smoking use
   
2.872
0.090
 None
65
60
5
  
 Yes
114
95
19
  
Alcohol use
   
0.637
0.425
 None
73
65
8
  
 Yes
106
90
16
  
Tumor location
   
0.771
0.680
 Upper
20
16
4
  
 Middle
97
85
12
  
 Lower
62
54
8
  
Histological grade
   
1.557
0.459
 I
32
29
3
  
 II
98
86
12
  
 III
49
40
9
  
Tumor invasion depth
   
7.935
0.019
 T1-T2
39
38
1
  
 T3
110
94
16
  
 T4a
30
23
7
  
Lymph node metastasis
   
10.840
0.013
 N0
83
76
7
  
 N1
62
56
6
  
 N2
22
15
7
  
 N3
12
8
4
  
TNM stage
   
8.878
0.012
 I
10
10
0
  
 II
77
72
5
  
 III
92
73
19
  
Adjuvant therapy
   
1.768
0.184
 None
45
42
3
  
 Yes
104
87
17
  
 unknown
30
    
Abbreviations: PPFIA1 PTPRF interacting protein alpha 1, ESCC Esophageal squamous cell carcinoma, TNM tumor-node-metastasis
We then analyzed PPFIA1 levels in the cDNA array data of 67 ESCC patients. The patients were grouped into a low-expression group (n = 34) and a high-expression group (n = 33) based on the median value of relative PPFIA1 expression. We found that PPFIA1 mRNA expression was significantly correlated with histological grade (P = 0.031, Supplementary Table 2).

Correlations between PPFIA1 protein expression and clinicopathological variables in ESCC patients in the TMA dataset

We further assessed PPFIA1 protein expression using an IHC staining-based TMA dataset containing samples from 147 surgically removed cancer tissues and 40 normal esophageal tissues. PPFIA1 was primarily localized to the cytoplasm and nucleus of cancer cells, and the expression rate was significantly higher in cancer tissues than in normal tissues (68.0% vs. 25.0%, P < 0.05). Representative images of low or high PPFIA1 expression are shown in Fig. 3A-C. All patients were divided into the PPFIA1 low-expressing group (n = 47) or the high-expressing group (n = 100). Correlation analysis found that PPFIA1 expression was inversely associated with tumor location (P = 0.011), tumor invasion depth (P = 0.041), lymph node metastasis (P = 0.020), and TNM stage (P = 0.007) in ESCC patients (Table 2).
Table 2
Correlations between PPFIA1 protein expression and clinicopathological variables in 147 patients with ESCC from the TMA dataset
Clinicopathological variables
Number
PPFIA1 expression
x2
P value
Low
High
Gender
   
< 0.001
0.983
 Male
119
38 (31.9%)
81 (68.1%)
  
 Female
28
9 (32.1%)
19 (67.9%)
  
Age(years)
   
1.433
0.231
 ≤ 68
77
28 (36.4%)
49 (63.6%)
  
 ˃68
70
19 (27.1%)
51 (72.9%)
  
Smoking use
   
3.280
0.070
 None
77
20 (26.0%)
57 (74.0%)
  
 Yes
70
28 (40.0%)
42 (60.0%)
  
Tumor location
   
9.067
0.011
 Upper
6
0 (0)
6 (100.0%)
  
 Middle
123
37 (30.1%)
86 (69.9%)
  
 Lower
18
10 (55.6%)
8 (44.4%)
  
Tumor size (cm)
   
0.803
0.370
 < 3.5
61
22 (36.1%)
39 (63.9%)
  
 ≥ 3.5
86
25 (29.1%)
61 (70.9%)
  
Histological grade
   
1.565
0.457
 I
11
5 (45.5%)
6 (54.5%)
  
 II
115
37 (32.2%)
78 (67.8%)
  
 III
21
5 (23.8%)
16 (76.2%)
  
Tumor invasion depth
   
6.369
0.041
 T1- T2
22
11 (50.0%)
11 (50.0%)
  
 T3
80
27 (33.8%)
53 (66.3%)
  
 T4
45
9 (20.0%)
36 (80.0%)
  
Lymph node metastasis
   
5.449
0.020
 None
86
34 (39.5%)
52 (60.5%)
  
 Yes
61
13 (21.3%)
48 (78.7%)
  
TNM stage
   
7.344
0.007
 I/II
55
25 (45.5%)
30 (54.5%)
  
 III
92
22 (23.9%)
70 (76.1%)
  
Abbreviations: PPFIA1 PTPRF interacting protein alpha 1, ESCC esophageal squamous cell carcinoma, TNM tumor-node-metastasis, TMA TMA, tissue microarray

Increased PPFIA1 expression indicates a poor prognosis for malignancies

In view of the high expression of PPFIA1 mRNA in a variety of malignancies, including breast, ovarian, lung, and gastric cancers, we further investigated the prognostic value of PPFIA1 through the Kaplan–Meier Plotter website. Patients were grouped based on the automatically selected best cutoff value for PPFIA1 expression. As shown in Supplementary Fig. 1, the expression of PPFIA1 apparently correlated with the RFS, OS, PPS, and DMFS of patients with breast cancer (P < 0.05); the PFS, OS, and PPS of patients with ovarian cancer (P < 0.05); the FP, OS, and PPS of patients with lung cancer (P < 0.05); and the FP, OS, and PPS of patients with gastric cancer (P < 0.05). These results suggest that high expression of PPFIA1 is obviously associated with worse outcomes in patients with breast cancer, ovarian cancer, lung cancer, and gastric cancer.

Correlations between PPFIA1 expression and the prognosis of ESCC patients

Survival analyses revealed that the median survival time of ESCC patients with high and low PPFIA1 expression were similar (22.7 months vs. 45.37 months, P = 0.349, Fig. 4A) in the TCGA database. ESCC patients with high PPFIA1 mRNA levels had a significantly worse 5-year OS rate than those with low PPFIA1 mRNA levels in the GSE53625 dataset (12.5% vs. 45.2%, P < 0.001, Fig. 4B) and cDNA array dataset (9.2% vs. 38.0%, P = 0.001, Fig. 4C). High PPFIA1 expression at the protein level was also obviously related to poor outcomes in patients in the TMA dataset (5-year OS rate: 23.4% vs. 46.0%, P = 0.002, Fig. 4D).
The prognosis-related indicators in the 3 datasets were analyzed by univariate and multivariate survival analyses. In the GSE53625 dataset, age, histological grade, lymph node metastasis, adjuvant therapy and PPFIA1 expression were significantly related to OS. In the cDNA array dataset, lymph node metastasis, M status, TNM stage and PPFIA1 expression were related to OS. Moreover, age, tumor size, histological grade, tumor invasion depth, lymph node metastasis, and PPFIA1 expression were significantly related to OS in the TMA dataset. Furthermore, PPFIA1 was identified as an independent risk factor for poor OS in the multivariate Cox proportional hazard regression analysis of all 3 datasets (GSE53625 dataset, HR = 1.942, P = 0.019, Fig. 5A; cDNA array dataset, HR = 3.464, P < 0.001, Fig. 5B; TMA dataset, HR = 1.596, P = 0.039, Fig. 5C). Other independent factors included tumor size (TMA dataset), lymph node metastasis (GSE53625 and TMA datasets), TNM stage (cDNA array dataset) and adjuvant therapy (GSE53625 dataset).

The effect of PPFIA1 on the migration and invasion of ESCC cells in vitro

To further determine the role of PPFIA1 in ESCC cell migration and invasion, we transfected Kyse-30 and Ec-109 cells with PPFIA1-siRNA or negative control. We found the PPFIA1 expression at the mRNA (Fig. 6A) and protein (Fig. 6B) levels were downregulated by transfection with PPFIA1-siRNA (P < 0.05). The wound-healing assay showed that PPFIA1 knockdown significantly inhibited the migration ability of Kyse-30 and Ec-109 cells (Fig. 6C, P < 0.05). Furthermore, transwell assays showed that the relative migration and invasion abilities of Kyse-30 and Ec-109 cells were remarkably decreased after PPFIA1 knockdown (Fig. 6D, P < 0.05). Thus, these results indicate that the expression of PPFIA1 is crucial for the migration and invasion of ESCC cells.

Network analysis of PPFIA1 and interacting genes

The interactions between PPFIA1 and other genes in the PPI network were analyzed via the STRING website. The results showed some known and predicted interactions between PPFIA1 and UNC13B, RAB3A, PTPRD, SYT1, RIMS1, PTPRS, APBA1, PTPRF, LIN7A, and CASK (Fig. 7A). Most of these genes have been reported as oncogenes or tumor suppressor genes.
We further analyzed the associations of PPFIA1 mRNA expression with the abovementioned genes in the GSE53625 dataset. Our results showed that PPFIA1 expression was clearly correlated with UNC13B, RAB3A, PTPRD, SYT1, RIMS1, PTPRS, APBA1, PTPRF, and LIN7A expression (Fig. 7B-J) but not with CASK expression (Fig. 7K).

Discussion

Early invasion and metastasis are two of the main reasons for the poor prognosis of patients with ESCC [19]. In our current study, we observed an increase of PPFIA1 expression in ESCC, which was associated with tumor metastasis and a poorer prognosis. PPFIA1 may be a potential molecular indicator for diagnosis and prognosis evaluation and could be regarded as a new therapeutic target for ESCC.
PPFIA1 is a gene located at the 11q13 amplification region and mainly encodes the liprin-α1 protein in humans. It was initially discovered to control the formation and function of synapses in neurons [20]. Studies have indicated that PPFIA1 is a key regulator of cell motility, focal adhesion, cell signal transduction, and cytoskeletal organization [21, 22]. PPFIA1 is frequently amplified and plays a crucial role in cell migration and invasion by affecting cell motility, mediating extracellular matrix degradation and facilitating the formation of lamellipodial protrusions of cancer cells [5, 23, 24]. Although the role of PPFIA1 in tumor cell progression has been well verified, it is not clear whether its dysregulation is related to metastasis risk or prognosis in ESCC patients.
To determine the clinical value of PPFIA1 in ESCC, we analyzed the difference in the expression of this novel marker in tumor tissues compared with normal tissues and assessed its correlation with the survival of ESCC patients using multiple datasets. Analysis of datasets from Oncomine, GEPIA and GEO confirmed that PPFIA1 mRNA expression was markedly higher in ESCC tissues than in adjacent normal control tissues, which was further confirmed with cDNA array data based on qRT–PCR and TMA data based on IHC. These results indicated that PPFIA1 may play a vital role during the tumorigenesis of ESCC, which is consistent with the results discovered in other malignant tumors [20, 23]. Further correlation analysis revealed that PPFIA1 expression is highly correlated with aggressive biological behaviors of tumors, indicating the important function of PPFIA1 in the progression of ESCC. It is worth mentioning that the expression of PPFIA1 was only correlated with the degree of tumor differentiation in the cDNA chip dataset, which might be due to the limited sample size in the research.
We further discovered that PPFIA1 can be applied for the prognostic assessment of breast cancer, ovarian cancer, lung cancer and gastric cancer by online Kaplan–Meier Plotter. Cho et al. [25]. explored the prognostic value of PPFIA1 alone or in combination with TMEM16A and FADD in patients with invasive breast cancer and found that combined expression was significantly associated with perineural invasion and a low disease-free survival rate. A recent study showed that the expression of PPFIA1 is closely related to poor response to endocrine treatment in luminal breast cancer [21]. Since PPFIA1 was found to be involved in the development of ESCC, it could be inferred that PPFIA1 is likely to have a great impact on the survival of patients with ESCC. To assess this possibility, we used the GSE53625, cDNA array and TMA datasets to investigate the prognostic value of PPFIA1 in ESCC. Our results indicated that high PPFIA1 expression was evidently correlated with a poorer prognosis. Notably, PPFIA1 was identified as an independent indicator of poor prognosis in all three independent databases in the multivariate analyses. Thus, PPFIA1 may serve as a potential diagnostic and novel prognostic biomarker, as well as a new therapeutic target for ESCC.
Since the expression of PPFIA1 is related to malignant biological indicators and poor prognosis, we further carried out cytological experiments to clarify the effect of PPFIA1 on the metastasis ability of ESCC cells. Our study found that down-regulation of PPFIA1 expression can significantly reduce the migration and invasion behavior of cancer cells. These data further support that PPFIA1 functions as an oncogene in the metastasis of ESCC.
Mechanistically, Shen et al. [26] confirmed that PPFIA1 enhances the proliferation and migration ability of colon carcinoma cells by interacting with the tumor suppressor protein ING4. Astro et al. [27] suggested that liprin-α1 can determine the polarization and morphological dynamics related to cell migration by forming a complex with the liprin-β1, ERC1/ELKS, and LL5 proteins. To examine the potential mechanisms of PPFIA1 in ESCC progression, we further used PPI network and Pearson’s correlation analyses to identify proteins that may bind to PPFIA1. We identified UNC13B, RAB3A, PTPRD, SYT1 and RIMS1 as potential candidates. Some of these interacting genes have already been reported as tumor oncogenes or suppressor genes. For instance, RAB3A interacting protein (Rab3IP) is a Rab-specific GEF, and the activation of Rab proteins, including RAB3A and RAB8, has been considered a tumor-specific marker in colorectal cancer, gastric cancer, and pancreatic cancer [28, 29]. A recent study reported by Ren et al. [30] showed that RAB3IP interacts with SSX2 and enhances the invasive aggressive phenotype of gastric cancer through epithelial-mesenchymal transition. PTPRT is a phosphatase that can participate in JAK/STAT signal transduction. Deleterious mutations or copy number loss of PTPRT and its related gene PTPRD are potential markers for evaluating resistance to bevacizumab regimens and are closely associated with shorter PFS in metastatic colorectal cancer patients [31]. However, the biological function and underlying mechanism of PPFIA1 in ESCC need to be further studied.
There are several certain limitations. First, although the results were analyzed through bioinformatics analysis and three independent databases (GSE53625, cDNA array and TMA), the sample size involved in the present study was relatively small. Second, the clinicopathological information from the three datasets was not comprehensive, and part of the information in the cDNA array dataset was incomplete. Third, the detailed molecular mechanism and signaling pathways by which PPFIA1 influences the migration and invasion of ESCC were not verified. Therefore, our results need to be further verified.
In conclusion, this study indicates that the expression of PPFIA1 is significantly increased and is related to migration and invasion behaviors, and poor outcomes in ESCC patients. PPFIA1 might be a valuable biomarker for early detection, treatment formulation and prognostic evaluation for ESCC.

Acknowledgements

Not Applicable.

Declarations

All methods were performed in accordance with the relevant ethical regulations and were conducted under approvals of the Research Ethics Committee of Tianjin Medical University Cancer Institute and Hospital. Informed consent was acquired from all patients of primary cohort for the acquisition of clinical and pathological information and the use of surgical specimens.
Not applicable.

Competing interests

The authors declare 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 Sung H, Ferlay J, Siegel RL, et al. Global Cancer Statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2021;71(3):209–49.CrossRefPubMed Sung H, Ferlay J, Siegel RL, et al. Global Cancer Statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2021;71(3):209–49.CrossRefPubMed
2.
Zurück zum Zitat Morgan E, Soerjomataram I, Rumgay H, et al. The global landscape of esophageal squamous cell carcinoma and esophageal adenocarcinoma incidence and mortality in 2020 and projections to 2040: new estimates from GLOBOCAN 2020. Gastroenterology. 2022;163(3):649-58.e2.CrossRefPubMed Morgan E, Soerjomataram I, Rumgay H, et al. The global landscape of esophageal squamous cell carcinoma and esophageal adenocarcinoma incidence and mortality in 2020 and projections to 2040: new estimates from GLOBOCAN 2020. Gastroenterology. 2022;163(3):649-58.e2.CrossRefPubMed
3.
Zurück zum Zitat Zhang H, Xiao W, Ren P, et al. The prognostic performance of the log odds of positive lymph nodes in patients with esophageal squamous cell carcinoma: a population study of the US SEER database and a Chinese single-institution cohort. Cancer Med. 2021;10(17):6149–64.CrossRefPubMedPubMedCentral Zhang H, Xiao W, Ren P, et al. The prognostic performance of the log odds of positive lymph nodes in patients with esophageal squamous cell carcinoma: a population study of the US SEER database and a Chinese single-institution cohort. Cancer Med. 2021;10(17):6149–64.CrossRefPubMedPubMedCentral
4.
Zurück zum Zitat Pehkonen H, von Nandelstadh P, Karhemo PR, et al. Liprin-α1 is a regulator of vimentin intermediate filament network in the cancer cell adhesion machinery. Sci Rep. 2016;6:24486.CrossRefPubMedPubMedCentral Pehkonen H, von Nandelstadh P, Karhemo PR, et al. Liprin-α1 is a regulator of vimentin intermediate filament network in the cancer cell adhesion machinery. Sci Rep. 2016;6:24486.CrossRefPubMedPubMedCentral
5.
Zurück zum Zitat Astro V, Asperti C, Cangi MG, Doglioni C, de Curtis I. Liprin-α1 regulates breast cancer cell invasion by affecting cell motility, invadopodia and extracellular matrix degradation. Oncogene. 2011;30(15):1841–9.CrossRefPubMed Astro V, Asperti C, Cangi MG, Doglioni C, de Curtis I. Liprin-α1 regulates breast cancer cell invasion by affecting cell motility, invadopodia and extracellular matrix degradation. Oncogene. 2011;30(15):1841–9.CrossRefPubMed
6.
Zurück zum Zitat Blessmann M, Al-Dam A, Hanken H, et al. Amplification of the PPFIA1 gene region on 11q13 in oral squamous cell carcinomas (OSCC). J Craniomaxillofac Surg. 2013;41(8):845–9.CrossRefPubMed Blessmann M, Al-Dam A, Hanken H, et al. Amplification of the PPFIA1 gene region on 11q13 in oral squamous cell carcinomas (OSCC). J Craniomaxillofac Surg. 2013;41(8):845–9.CrossRefPubMed
7.
Zurück zum Zitat Brown LA, Kalloger SE, Miller MA, et al. Amplification of 11q13 in ovarian carcinoma. Genes Chromosomes Cancer. 2008;47(6):481–9.CrossRefPubMed Brown LA, Kalloger SE, Miller MA, et al. Amplification of 11q13 in ovarian carcinoma. Genes Chromosomes Cancer. 2008;47(6):481–9.CrossRefPubMed
8.
Zurück zum Zitat Chu J, Min KW, Kim DH, et al. High PPFIA1 expression promotes cancer survival by suppressing CD8+ T cells in breast cancer: drug discovery and machine learning approach. Breast Cancer. 2023;30(2):259–70.CrossRefPubMed Chu J, Min KW, Kim DH, et al. High PPFIA1 expression promotes cancer survival by suppressing CD8+ T cells in breast cancer: drug discovery and machine learning approach. Breast Cancer. 2023;30(2):259–70.CrossRefPubMed
9.
Zurück zum Zitat Fu F, Niu R, Zheng M, et al. Clinicopathological significances and prognostic value of PPFIA4 in colorectal cancer. J Cancer. 2023;14(1):24–34.CrossRefPubMedPubMedCentral Fu F, Niu R, Zheng M, et al. Clinicopathological significances and prognostic value of PPFIA4 in colorectal cancer. J Cancer. 2023;14(1):24–34.CrossRefPubMedPubMedCentral
10.
Zurück zum Zitat Zhou H, Cao T, Li WP, Wu G. Combined expression and prognostic significance of PPFIA1 and ALG3 in head and neck squamous cell carcinoma. Mol Biol Rep. 2019;46(3):2693–701.CrossRefPubMed Zhou H, Cao T, Li WP, Wu G. Combined expression and prognostic significance of PPFIA1 and ALG3 in head and neck squamous cell carcinoma. Mol Biol Rep. 2019;46(3):2693–701.CrossRefPubMed
11.
Zurück zum Zitat Tang Z, Li C, Kang B, Gao G, Li C, Zhang Z. GEPIA: a web server for cancer and normal gene expression profiling and interactive analyses. Nucleic Acids Res. 2017;45(W1):W98-98W102.CrossRefPubMedPubMedCentral Tang Z, Li C, Kang B, Gao G, Li C, Zhang Z. GEPIA: a web server for cancer and normal gene expression profiling and interactive analyses. Nucleic Acids Res. 2017;45(W1):W98-98W102.CrossRefPubMedPubMedCentral
12.
Zurück zum Zitat Rhodes DR, Kalyana-Sundaram S, Mahavisno V, et al. Oncomine 3.0: genes, pathways, and networks in a collection of 18,000 cancer gene expression profiles. Neoplasia. 2007;9(2):166–80.CrossRefPubMedPubMedCentral Rhodes DR, Kalyana-Sundaram S, Mahavisno V, et al. Oncomine 3.0: genes, pathways, and networks in a collection of 18,000 cancer gene expression profiles. Neoplasia. 2007;9(2):166–80.CrossRefPubMedPubMedCentral
13.
Zurück zum Zitat Yu-Jing T, Wen-Jing T, Biao T. Integrated analysis of hub genes and pathways in esophageal carcinoma based on NCBI’s Gene Expression Omnibus (GEO) database: a bioinformatics analysis. Med Sci Monit. 2020;26:e923934.CrossRefPubMedPubMedCentral Yu-Jing T, Wen-Jing T, Biao T. Integrated analysis of hub genes and pathways in esophageal carcinoma based on NCBI’s Gene Expression Omnibus (GEO) database: a bioinformatics analysis. Med Sci Monit. 2020;26:e923934.CrossRefPubMedPubMedCentral
14.
Zurück zum Zitat Xiao W, Tang P, Sui Z, et al. Establishment of a risk model by integrating hypoxia genes in predicting prognosis of esophageal squamous cell carcinoma. Cancer Med. 2023;12(2):2117–33.CrossRefPubMed Xiao W, Tang P, Sui Z, et al. Establishment of a risk model by integrating hypoxia genes in predicting prognosis of esophageal squamous cell carcinoma. Cancer Med. 2023;12(2):2117–33.CrossRefPubMed
15.
Zurück zum Zitat Alatan H, Chen Y, Zhou J, Wang L. Extracellular matrix-related hubs genes have adverse effects on gastric adenocarcinoma prognosis based on bioinformatics analysis. Genes (Basel). 2021;12(7):1104.CrossRefPubMed Alatan H, Chen Y, Zhou J, Wang L. Extracellular matrix-related hubs genes have adverse effects on gastric adenocarcinoma prognosis based on bioinformatics analysis. Genes (Basel). 2021;12(7):1104.CrossRefPubMed
16.
Zurück zum Zitat Zhang H, Wu X, Sui Z, et al. High-mobility group AT-hook 2 promotes growth and metastasis and is regulated by miR-204-5p in oesophageal squamous cell carcinoma. Eur J Clin Invest. 2021;51(8):e13563.CrossRefPubMed Zhang H, Wu X, Sui Z, et al. High-mobility group AT-hook 2 promotes growth and metastasis and is regulated by miR-204-5p in oesophageal squamous cell carcinoma. Eur J Clin Invest. 2021;51(8):e13563.CrossRefPubMed
17.
Zurück zum Zitat Wu X, Zhang H, Sui Z, et al. CXCR4 promotes the growth and metastasis of esophageal squamous cell carcinoma as a critical downstream mediator of HIF-1α. Cancer Sci. 2022;113(3):926–39.CrossRefPubMedPubMedCentral Wu X, Zhang H, Sui Z, et al. CXCR4 promotes the growth and metastasis of esophageal squamous cell carcinoma as a critical downstream mediator of HIF-1α. Cancer Sci. 2022;113(3):926–39.CrossRefPubMedPubMedCentral
18.
Zurück zum Zitat Ogłuszka M, Orzechowska M, Jędroszka D, Witas P, Bednarek AK. Evaluate cutpoints: adaptable continuous data distribution system for determining survival in Kaplan-Meier estimator. Comput Methods Programs Biomed. 2019;177:133–9.CrossRefPubMed Ogłuszka M, Orzechowska M, Jędroszka D, Witas P, Bednarek AK. Evaluate cutpoints: adaptable continuous data distribution system for determining survival in Kaplan-Meier estimator. Comput Methods Programs Biomed. 2019;177:133–9.CrossRefPubMed
19.
Zurück zum Zitat Zhang J, Luo A, Huang F, Gong T, Liu Z. SERPINE2 promotes esophageal squamous cell carcinoma metastasis by activating BMP4. Cancer Lett. 2020;469:390–8.CrossRefPubMed Zhang J, Luo A, Huang F, Gong T, Liu Z. SERPINE2 promotes esophageal squamous cell carcinoma metastasis by activating BMP4. Cancer Lett. 2020;469:390–8.CrossRefPubMed
20.
Zurück zum Zitat Barros-Filho MC, Reis-Rosa LA, Hatakeyama M, et al. Oncogenic drivers in 11q13 associated with prognosis and response to therapy in advanced oropharyngeal carcinomas. Oral Oncol. 2018;83:81–90.CrossRefPubMed Barros-Filho MC, Reis-Rosa LA, Hatakeyama M, et al. Oncogenic drivers in 11q13 associated with prognosis and response to therapy in advanced oropharyngeal carcinomas. Oral Oncol. 2018;83:81–90.CrossRefPubMed
21.
Zurück zum Zitat Alfarsi LH, El Ansari R, Craze ML, et al. PPFIA1 expression associates with poor response to endocrine treatment in luminal breast cancer. BMC Cancer. 2020;20(1):425.CrossRefPubMedPubMedCentral Alfarsi LH, El Ansari R, Craze ML, et al. PPFIA1 expression associates with poor response to endocrine treatment in luminal breast cancer. BMC Cancer. 2020;20(1):425.CrossRefPubMedPubMedCentral
22.
Zurück zum Zitat Dancau AM, Wuth L, Waschow M, et al. PPFIA1 and CCND1 are frequently coamplified in breast cancer. Genes Chromosomes Cancer. 2010;49(1):1–8.CrossRefPubMed Dancau AM, Wuth L, Waschow M, et al. PPFIA1 and CCND1 are frequently coamplified in breast cancer. Genes Chromosomes Cancer. 2010;49(1):1–8.CrossRefPubMed
23.
Zurück zum Zitat Chiaretti S, de Curtis I. Role of Liprins in the regulation of tumor cell motility and invasion. Curr Cancer Drug Targets. 2016;16(3):238–48.CrossRefPubMed Chiaretti S, de Curtis I. Role of Liprins in the regulation of tumor cell motility and invasion. Curr Cancer Drug Targets. 2016;16(3):238–48.CrossRefPubMed
24.
Zurück zum Zitat Yang J, Wu NN, Huang DJ, et al. PPFIA1 is upregulated in liver metastasis of breast cancer and is a potential poor prognostic indicator of metastatic relapse. Tumour Biol. 2017;39(7):1010428317713492.CrossRefPubMed Yang J, Wu NN, Huang DJ, et al. PPFIA1 is upregulated in liver metastasis of breast cancer and is a potential poor prognostic indicator of metastatic relapse. Tumour Biol. 2017;39(7):1010428317713492.CrossRefPubMed
25.
Zurück zum Zitat Choi EJ, Yun JA, Jabeen S, et al. Prognostic significance of TMEM16A, PPFIA1, and FADD expression in invasive ductal carcinoma of the breast. World J Surg Oncol. 2014;12:137.CrossRefPubMedPubMedCentral Choi EJ, Yun JA, Jabeen S, et al. Prognostic significance of TMEM16A, PPFIA1, and FADD expression in invasive ductal carcinoma of the breast. World J Surg Oncol. 2014;12:137.CrossRefPubMedPubMedCentral
26.
Zurück zum Zitat Shen JC, Unoki M, Ythier D, et al. Inhibitor of growth 4 suppresses cell spreading and cell migration by interacting with a novel binding partner, liprin alpha1. Cancer Res. 2007;67(6):2552–8.CrossRefPubMedPubMedCentral Shen JC, Unoki M, Ythier D, et al. Inhibitor of growth 4 suppresses cell spreading and cell migration by interacting with a novel binding partner, liprin alpha1. Cancer Res. 2007;67(6):2552–8.CrossRefPubMedPubMedCentral
27.
Zurück zum Zitat Astro V, Chiaretti S, Magistrati E, Fivaz M, de Curtis I. Liprin-α1, ERC1 and LL5 define polarized and dynamic structures that are implicated in cell migration. J Cell Sci. 2014;127(Pt 17):3862–76.PubMed Astro V, Chiaretti S, Magistrati E, Fivaz M, de Curtis I. Liprin-α1, ERC1 and LL5 define polarized and dynamic structures that are implicated in cell migration. J Cell Sci. 2014;127(Pt 17):3862–76.PubMed
28.
Zurück zum Zitat Guo W, Chen Z, Chen Z, et al. Promotion of Cell Proliferation through inhibition of cell autophagy signalling pathway by Rab3IP is restrained by MicroRNA-532-3p in gastric cancer. J Cancer. 2018;9(23):4363–73.CrossRefPubMedPubMedCentral Guo W, Chen Z, Chen Z, et al. Promotion of Cell Proliferation through inhibition of cell autophagy signalling pathway by Rab3IP is restrained by MicroRNA-532-3p in gastric cancer. J Cancer. 2018;9(23):4363–73.CrossRefPubMedPubMedCentral
29.
Zurück zum Zitat Hur K, Cejas P, Feliu J, et al. Hypomethylation of long interspersed nuclear element-1 (LINE-1) leads to activation of proto-oncogenes in human colorectal cancer metastasis. Gut. 2014;63(4):635–46.CrossRefPubMed Hur K, Cejas P, Feliu J, et al. Hypomethylation of long interspersed nuclear element-1 (LINE-1) leads to activation of proto-oncogenes in human colorectal cancer metastasis. Gut. 2014;63(4):635–46.CrossRefPubMed
30.
Zurück zum Zitat Ren H, Xu Z, Guo W, Deng Z, Yu X. Rab3IP interacts with SSX2 and enhances the invasiveness of gastric cancer cells. Biochem Biophys Res Commun. 2018;503(4):2563–8.CrossRefPubMed Ren H, Xu Z, Guo W, Deng Z, Yu X. Rab3IP interacts with SSX2 and enhances the invasiveness of gastric cancer cells. Biochem Biophys Res Commun. 2018;503(4):2563–8.CrossRefPubMed
31.
Zurück zum Zitat Hsu HC, Lapke N, Chen SJ, et al. PTPRT and PTPRD deleterious mutations and deletion predict bevacizumab resistance in metastatic colorectal cancer patients. Cancers (Basel). 2018;10(9):314.CrossRefPubMed Hsu HC, Lapke N, Chen SJ, et al. PTPRT and PTPRD deleterious mutations and deletion predict bevacizumab resistance in metastatic colorectal cancer patients. Cancers (Basel). 2018;10(9):314.CrossRefPubMed
Metadaten
Titel
High expression of PPFIA1 in human esophageal squamous cell carcinoma correlates with tumor metastasis and poor prognosis
verfasst von
Yongyin Gao
Lizhao Guan
Ran Jia
Wanyi Xiao
Youming Han
Yue Li
Peng Tang
Zhentao Yu
Hongdian Zhang
Publikationsdatum
01.12.2023
Verlag
BioMed Central
Erschienen in
BMC Cancer / Ausgabe 1/2023
Elektronische ISSN: 1471-2407
DOI
https://doi.org/10.1186/s12885-023-10872-9

Weitere Artikel der Ausgabe 1/2023

BMC Cancer 1/2023 Zur Ausgabe

Blutdrucksenkung könnte Uterusmyome verhindern

Frauen mit unbehandelter oder neu auftretender Hypertonie haben ein deutlich erhöhtes Risiko für Uterusmyome. Eine Therapie mit Antihypertensiva geht hingegen mit einer verringerten Inzidenz der gutartigen Tumoren einher.

Alphablocker schützt vor Miktionsproblemen nach der Biopsie

16.05.2024 alpha-1-Rezeptorantagonisten Nachrichten

Nach einer Prostatabiopsie treten häufig Probleme beim Wasserlassen auf. Ob sich das durch den periinterventionellen Einsatz von Alphablockern verhindern lässt, haben australische Mediziner im Zuge einer Metaanalyse untersucht.

Antikörper-Wirkstoff-Konjugat hält solide Tumoren in Schach

16.05.2024 Zielgerichtete Therapie Nachrichten

Trastuzumab deruxtecan scheint auch jenseits von Lungenkrebs gut gegen solide Tumoren mit HER2-Mutationen zu wirken. Dafür sprechen die Daten einer offenen Pan-Tumor-Studie.

Mammakarzinom: Senken Statine das krebsbedingte Sterberisiko?

15.05.2024 Mammakarzinom Nachrichten

Frauen mit lokalem oder metastasiertem Brustkrebs, die Statine einnehmen, haben eine niedrigere krebsspezifische Mortalität als Patientinnen, die dies nicht tun, legen neue Daten aus den USA nahe.

Update Onkologie

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