Skip to main content
Erschienen in: Molecular Cancer 1/2018

Open Access 01.12.2018 | Research

Proteogenomic characterization and comprehensive integrative genomic analysis of human colorectal cancer liver metastasis

verfasst von: Yu-Shui Ma, Tao Huang, Xiao-Ming Zhong, Hong-Wei Zhang, Xian-Ling Cong, Hong Xu, Gai-Xia Lu, Fei Yu, Shao-Bo Xue, Zhong-Wei Lv, Da Fu

Erschienen in: Molecular Cancer | Ausgabe 1/2018

Abstract

Background

Proteogenomic characterization and integrative and comparative genomic analysis provide a functional context to annotate genomic abnormalities with prognostic value.

Methods

Here, we analyzed the proteomes and performed whole exome and transcriptome sequencing and single nucleotide polymorphism array profiling for 2 sets of triplet samples comprised of normal colorectal tissue, primary CRC tissue, and synchronous matched liver metastatic tissue.

Results

We identified 112 CNV-mRNA-protein correlated molecules, including up-regulated COL1A2 and BGN associated with prognosis, and four strongest hot spots (chromosomes X, 7, 16 and 1) driving global mRNA abundance variation in CRC liver metastasis. Two sites (DMRTB1R202H and PARP4V458I) were revealed to frequent mutate only in the liver metastatic cohort and displayed dysregulated protein abundance. Moreover, we confirmed that the mutated peptide number has potential prognosis value and somatic variants displayed increased protein abundance, including high MYH9 and CCT6A expression, with clinical significance.

Conclusions

Our proteogenomic characterization and integrative and comparative genomic analysis provides a new paradigm for understanding human colon and rectal cancer liver metastasis.

Trial registration

ClinicalTrials, NCT02917707. Registered 28 September 2016, https://​clinicaltrials.​gov/​ct2/​show/​NCT02917707.
Hinweise
Yu-Shui Ma, Tao Huang, Xiao-Ming Zhong and Hong-Wei Zhang contributed equally to this work.
A correction to this article is available online at https://​doi.​org/​10.​1186/​s12943-019-1005-3.
Abkürzungen
CLM
Colorectal cancer liver metastasis
CNV
Copy number variation
CRC
Colorectal cancer
DEmRNAs
Differentially expressed mRNAs
DEproteins
Differential expression proteins
LC-MS/MS
Liquid chromatography-tandem mass spectrometry
LM
Synchronous matched liver metastatic tissue from CRC patients
MT
Primary CRC tumor tissue from CRC patients with liver metastasis
NM
Primary CRC tissue from CRC patients without liver metastasis
PN
Para-tumor normal colorectal tissue
SAAVs
Single amino acid variants
TCGA
The Cancer Genome Atlas

Background

Colorectal cancer (CRC) is a significant contributor of cancer morbidity and mortality [1]. Almost half of CRC patients die within 5 years of diagnosis due to the development of recurrent disease and metastasis [2]. Therefore, it is important to illuminate the molecular basis of CRC liver metastasis (CLM) in hopes of developing new effective treatment modalities.
The Cancer Genome Atlas (TCGA) has characterized the genomic features of many types of human cancers, including CRC [35] and The Clinical Proteomic Tumor Analysis Consortium has also performed CRC-integrated proteomic analyses [6]. However, the primary genetic basis of CLM has not been fully elucidated. Understanding the genetic and proteogenomic differences between primary colon cancer and associated metastases to the liver is essential for discovering metastasis-specific molecular biomarkers and for devising a better therapeutic approach for this disease.
In the present work, we report a comprehensive molecular characterization of human CLM. Multi-platform integration revealed that CRC metastatic to the liver is driven by diverse alterations affecting multiple genes and pathways. Proteogenomic characterization and integrative and comparative genomic analysis provides a functional context to annotate genomic abnormalities with prognostic value, as well as a new paradigm for understanding human colon and rectal cancer liver metastasis.

Methods

Patient specimen acquisition

The study was examined and approved by the Ethics Committee of the Shanghai Tenth People’s Hospital, Tongji University School of Medicine (SHSY-IEC-PAP-16-24). This study was registered with ClinicalTrials.​gov, number NCT02917707. Each participant provided their written informed consent to participate in this study. The inclusion criteria included: age ≤ 75 years with histologically proven CRC, no severe major organ dysfunction, WHO performance status of 0 or 1, or no prior cancer chemotherapy. The exclusion criteria included: age ≥ 76, severe major organ dysfunction, World Health Organization (WHO) performance status of > 1, or prior cancer chemotherapy. The morphology of primary CRC and paracarcinoma normal colorectal tissues was confirmed by two independent pathologists using cryostat frozen sections stained with hematoxylin and eosin. Follow-up data and statistics were recorded for all patients through Dec. 31, 2017.

DNA and RNA extraction

Using a co-isolation protocol, DNA and RNA were purified simultaneously using the QIAGEN All Prep DNA/RNA Micro Kit (Qiagen, CA, USA) according to the manufacturer’s instructions. The nucleic acid concentration was determined using a Nanodrop1000 spectrophotometer (Thermo Fisher Scientific; Waltham, MA, USA), and the RNA purity was verified using 1.5% denaturing agarose gels.

Protein extraction and analysis by LC-MS/MS

Fresh CRC tissues and para-tumor normal colorectal tissues (PN) were used for proteogenomic analysis. Three different parts of the same lesions for every sample were compared for data analysis and measurement of the variation caused by random biological effects. The samples were cut into small pieces (about 1 mm3) and rinsed in PBS to remove the blood. Then the tissues were homogenized in 4% SDS and 0.1 M DTT in 0.1 M Tris-HCl, pH 7.6 on ice, sonicated 10 times (80 w; 10 s sonication/5 s suspension), incubated for 3 min at 95 °C, and briefly sonicated. The protein concentrations of clarified lysates were determined using a fluorescence assay and then 200 μg of clarified lysates were proteolyzed on a 10 kDa filter (PALL Life Sciences, Shanghai, China) using the filter-aided sample preparation method [7]. The peptide samples were then desalted onto a solid-phase extraction cartridge. The lyophilized peptide mixture was re-suspended in water with 0.1% formic acid (v/v), and its content was estimated by ultraviolet light spectral density at 280 nm [8]. Then, 3 μg of the digest sample was analyzed by nano-liquid chromatography-tandem mass spectrometry on a LTQ Orbitrap Velos Pro mass spectrometer as previously described [9].
The acquired data from mass spectrometry runs were combined and searched against the UniProt Human database (05/2016, 153,652 entrys) using Maxquant software (version 1.3.0.5; http://​maxquant.​org/) as described [10]. Proteins were identified using the Andromeda peptide search engine integrated into the Maxquant platform. Trypsin-digested fragments were analyzed, allowing for a maximum of 2 missed cleavages. Carbamidomethyl cysteine was set as a fixed modification, with protein N-acetylation and methionine oxidation as variable modifications. Precursor ion tolerances were 20 ppm for first search and 6 ppm for a second search. The MS/MS peaks were de-isotoped and searched using a 20 ppm mass tolerance. The required minimum peptide length for identification was 7 amino acids, and the false discovery rate at the protein level, peptide level and site were set to 0.01. The normalized spectral protein intensity (label-free quantification) values were calculated for each protein group.
The Maxquant peptide and protein quantification result files were imported into Perseus software (version 1.5.1.6) to identify the differentially expressed proteins. After importing the quantitative data from ProteinGroups.txt into Perseus, a filtering criterion is set to keep the identified proteins with the quantified values of all ten reporter ions (no missing value) in the final identification list. The protein intensities are log2-transformed and normalized by subtracting the median intensity in each column/sample. Principal component analysis is performed based on protein intensities to differentiate groups. Two-samples tests coupled with Benjamini–Hochberg (FDR cutoff of 0.05) correction are performed to identify the differentially expressed proteins.

RNA sequencing analysis

Six specimens from 2 CRC patients with metastasis (comprised of triplet sets of PN, primary CRC tumor samples with liver metastasis (MT), and synchronously matched liver metastasis focus tissues (LM)) and 3 specimens from 3 CRC patients without liver metastasis (NM) were obtained for RNA sequencing analysis. The mRNA libraries were separately generated from total RNA and constructed according to the standard Illumina RNA library preparation protocol (Illumina Inc., USA). Sequencing was performed on the Illumina Nextseq 500 platform according to the manufacturer’s instructions. Images generated by Nextseq 500 were converted into nucleotide sequences using a base call pipeline and stored in FASTQ format, and the raw reads were filtered prior to analyzing the data. Clean reads were mapped to reference Homo sapiens transcriptome sequences from the UCSC website (hg19) using Bowtie2 and Tophat 2.0.1 software. To annotate gene expression, reads per kilobases per million read values of each gene were calculated, and differentially expressed genes were extracted using this value. The formula for calculating these values was defined as: reads per kilobases per million read values = total exon reads / (mapped reads [millions] × exon length [kbp]).

Chromosome microarray analysis and whole exome sequencing

Six specimens from 2 patients (including 2 triplet sets of primary MT, matched CLM and PN) were used for chromosome microarray analysis and whole exome sequencing analysis. DNAs and cRNAs were hybridized to the Affymetrix CytoScan HD Array as described and recurrent genomic regions with DNA copy gain and loss were identified using GISTIC, version 2.0 [11]. Genomic DNA was enriched for exonic regions using the SureSelect Biotinylated RNA Library. The sequences of captured libraries were generated as 90-bp pair-end reads on an Illumina Hiseq2000. Sequencing reads were processed and mapped to the reference GRCh37/hg19 human genome assembly using the Burrows-Wheeler Aligner as described [12]. Further processing, including duplicate removal, local realignment, base quality recalibration, and filtering, as well as the identification of SNVs and indels, was performed using the Genome Analysis Toolkit [13], SAM [14], and Picard tools (http://​picard.​sourceforge.​net). Then, filters were applied to obtain higher confidence, and annotation and classification were performed using ANNOVAR [15]. The variant collection was excluded from positions reported in the 1000 Genomes Project and dbSNP. The mean sequencing depth in the target regions was 80.28× (range 71.5 to 92.85).

Validation of point mutations by PCR and sanger sequencing

The reliability of the exome analysis and somatic variant identification strategies was assessed using PCR and Sanger sequencing. PCR was performed using the GeneAmp PCR System 9700 (Applied Biosystems, Foster City, CA, USA). About 20 ng template DNA from each sample was used per reaction. The products were sequenced, and all sequences were analyzed with the Sequencing Analysis Software Version 5.2 (Applied Biosystems).

Assay design, PCR amplification and genotyping

A panel comprising 120 positive sites identified by Sanger sequencing were selected. These single nucleotide polymorphisms were located within genes of different functional categories. For the PCR amplification and single base extension reaction, primer pairs and extension primers were designed using Assay design suite v2.0. These primers were multiplexed and genotyped using the Sequenom MassARRAY platform integrating the iPLEXSBE reaction and MassARRAY technology (Agena Bioscience, San Diego, CA, USA) based on the MALDI-TOF MS assay [16].

Hierarchical clustering, gene ontology (GO) and Kyoto encyclopedia of genes and genomes (KEGG) pathway analysis

Hierarchical clustering was performed using MEV software (http://​mev.​tm4.​org/​, v4.7.0, TIGR). The matrix was presented graphically by colouring each expression result on the basis of measured colour range: lower limit ‘0.0’ was coloured green, upper limit ‘369.5’ was coloured red and midpoint value ‘37.5’ was coloured black. Pearson correlation was used as distance metric and the complete linkage method was used. To identify genes/proteins that are specifically dysregulated in CLM, we fixed the cutoff at 2-fold with a P value less than 0.05. Dysregulated genes/proteins were subjected to GO analysis and KEGG pathway analysis by DAVID (http://​david.​ncifcrf.​gov). Pathway analysis is used to map genes to KEGG pathways. The P value denotes the significance of the pathway correlations (P value < 0.05 is recommended).

TCGA data acquisition and processing

We downloaded RNA-sequencing data from 379 CRC patients from TCGA portal (https://​cancergenome.​nih.​gov/), 12 of which had liver metastasis at the time of diagnosis or during the five-year follow-up period, and 367 of which had CRC without metastasis to the liver. The mRNA expression levels were investigated in 379 CRC tissues and 32 PN tissues in TCGA datasets by Illumina HiSeq 2000 RNA Sequencing Version 2 analysis and normalized by the RSEM algorithm. Whole-exome sequencing mutation datasets were downloaded from TCGA data set to create a customized CRC mutation database. The clinical information recorded, including the patient’s characteristics, tumor characteristics, and overall and progression-free survival was assessed.

Cell lines and transfection

Human CRC cell line SW480 were purchased from the Cell Bank of the Chinese Academy of Sciences (Shanghai, China) and cultured in DMEM media (Invitrogen, Carlsbad, USA) and supplemented with 10% (v/v) fetal bovine serum, 100 U/ml penicillin, and 100 mg/ml streptomycin. SW480 cell lines were routinely tested for mycoplasma contamination, and have been authenticated with short-tandem repeat analysis. Cell culture was conducted at 37 °C in a humidified 5% CO2 incubator. For COL1A2 and BGN over-expression, the human full length cDNA with or without point mutation were cloned into the pMSCV-hygro vector. The SW480 cells with stable over-expression were polyclonal derivatives with hygromycin selection to avoid clonal variations in functional assays.

Scratch-wound assay

The human CRC SW480 cells were conducted at 37 °C in a humidified 5% CO2 incubator and cells were grown into confluency in 6-well plates. The monolayer was artificially injured by scratching across the plate with a 200 ul pipette tip. The wells were washed 3 times to remove detached cells or cell debris. After 12 h, digital images were captured using a camera-equipped, inverted microscope (Carl Zeiss, Inc., Thorwood, NY, USA) and wound width measurements were subtracted from wound width at time zero to obtain the net wound closure.

In vitro invasion assays

Corning Costar Transwell 24-well plates with 8-um-pore-size polycarbonate membrane filters (Costar, Cambridge, MA) coated with BD Matrigel matrix (Becton Dickinson, Bedford, MA) were maintained for 1 h at 37 °C, followed by the addition of 1 × 105 transfected cells suspended in 200 μl medium with 1% serum into the top of each well insert. Normal growth medium was added to the bottom wells. The cells were allowed to migrate for 24 h at 37 °C. The migrated cells were fixed with 10% methanol for 15 min. The invading cells on the lower surface of the membrane were stained with 0.5% crystal violet for 5 min at room temperature. Random fields were photographed and the stained cells were counted under a microscope (Nikon Corporation).

Statistical analysis

Data were expressed as means ± standard deviations. Categorical data were reported as numbers and percentages. F tests were used to assess the equality of variances for comparable groups. Paired t test, One-way analysis of variance (ANOVA), Kruskal-Wallis test, and χ2 tests were used to analyze mRNA expression. Forty four paired fresh CRC and PN tissues were used for survival analysis (Table 1). OS was measured from the date the patient underwent surgery until the date of death resulting from any cause or last known follow-up for patients still alive. DFS analysis was measured from the date the patient underwent surgery to the date of disease recurrence, death from any cause (ie, noncancer deaths were not censored), or until last contact with the patient. For time-to-event analyses, survival estimates were calculated by the Kaplan-Meier analysis, and groups were compared with the log-rank test. Clinical variables that were considered for single variable analyses were previously identified as confounding variables with impact on the prognosis of patients with colorectal cancer: age at diagnosis (continuous), sex, primary site (colon vs. rectum), pathological differentiation (well to moderate vs. poor), completeness of colorectal resection (R0 vs. R1), tumor size (≥ 5 cm vs. < 5 cm), number of primary foci (multiple vs. single) and necrosis (yes vs. no). The Spearman’s correlation coefficient was used to test the relationship of two independent groups. To identify genes/proteins that are specifically dysregulated in CLM, we fixed the cutoff at 2-fold with a P value less than 0.05. All calculations were performed with SPSS 20.0 software (SPSS Inc., Chicago, IL, USA).
Table 1
Summary of colorectal cancer patients demographic and clinical characteristics (N = 44)
Factor
Variables
Non-metastatic (N = 21)
Metastatic to liver (N = 23)
Number (%)
Number (%)
Age
≥ 60
14 (66.7%)
11 (47.8%)
<  60
7 (33.3%)
12 (52.2%)
Gender
Male
11 (52.4%)
10 (43.5%)
Female
10 (47.6%)
13 (56.5%)
Primary site
Colon
9 (42.9%)
14 (60.9%)
Rectum
12 (57.1%)
9 (39.1%)
Differentiation
Well
0 (0.0%)
0 (0.0%)
Moderately
16 (76.2%)
16 (69.6%)
Poorly
5 (23.8%)
7 (30.4%)
Completeness of colorectal resection
R0
21 (100.0%)
23 (100.0%)
R1
0 (0.0%)
0 (0.0%)
Diameter
≥ 5 cm
10 (47.6%)
10 (43.5%)
<  5 cm
11 (52.4%)
13 (56.5%)
Number of foci
Multiple
8 (38.1%)
9 (39.1%)
Single
13 (61.9%)
14 (60.9%)
TNM stage
I-II
5 (23.8%)
5 (21.7%)
III-IV
16 (76.2%)
18 (78.3%)
Necrosis
Yes
11 (52.4%)
10 (43.5%)
No
10 (47.6%)
13 (56.5%)

Results

Identification of peptides and proteins associated with CLM

Genomic features and proteomic analyses of CRC have been characterized; however, the primary genetic basis of CLM has not been fully elucidated, which is essential for discovering metastasis-specific molecular biomarkers and for devising a better therapeutic approach for this disease. To address these issues, we performed a nano-liquid chromatography-tandem mass spectrometry (LC-MS/MS)-based shotgun proteomics profiling of 2 sets of triplet samples comprised of para-tumor normal colorectal tissue (PN), primary CRC tissue (MT), and synchronous matched liver metastatic tissue (LM) (Fig. 1). Three different parts of the same lesions for every sample were compared for data analysis and measurement of the variation caused by random biological effects. A total of 596,234 spectra were used in the Andromeda engine search, and 26,375 unique peptides were identified in an assembly of 4198 protein groups with a protein-level false discovery rate of 1.0%. Ingenuity pathway analysis with all 4198 identified proteins showed that about 51% of the proteins were from the cytoplasm, 26% were from the nucleus, 9% were from the plasma membrane and 5% were from the extracellular space, whereas 9% of proteins remained unclassified (Fig. 2a). The random predicted cellular distribution of the proteins supports the quality of the sample preparation.
A scatter plot of protein abundance (label-free quantification intensity) between CRC and PN tissues showed that there was a great variation between the MT or LM tumors and PN tissue (Fig. 2b and c). However, the protein expression between the MT and LM group was positively correlated (R2 = 0.78) (Fig. 2d). These results suggest that liver metastasis focus and primary focus share similar protein profiles and that there are common molecular alterations at each stage of tumor development.

Identification of significantly dysregulated proteins in CLM

To identify proteins that are specifically dysregulated in CLM, we fixed the cutoff at 2-fold with a P value less than 0.05. Among the 4198 proteins, a total of 1041 proteins were significantly altered between MT and PN tissue, 636 (61.09%) of which were down-regulated and 405 (38.91%) of which were up-regulated (Fig. 2a, left bars). There were 754 proteins with significantly difference in LM tissues when compared with PN tissues and 632 proteins with significantly difference in LM tissues when compared with MT tissues (Fig. 2d). Among that, 656 (47.33%) of which were down-regulated and 730 (52.67%) of which were up-regulated (Fig. 2a, middle and right bars) and 198 significantly differential expression proteins (DEproteins) (14.28%) involved in metabolic pathways (P = 3.28E-14) (Fig. 2e). These results suggest that metabolism-related pathways may play important roles in the liver metastasis of CRC.
To explore the functions of proteins that are dysregulated in CLM, we used DAVID analysis software to classify the Gene Ontology of the 1386 significantly altered proteins in LM tissues according to their molecular functions and cellular components and ranked them according to their biological processes (Fig. 2f). The top-ranked biological function was metabolites and energy, organonitrogen compound, cellular respiration, ATP metabolic process, localization, which suggests that metabolism-related biological function is associated with CLM (Fig. 2b).

Identification of significantly dysregulated mRNAs in CLM

Next, we performed RNA sequencing to identify differentially expressed mRNAs (DEmRNAs) that are specifically dysregulated in CLM. Unsupervised hierarchical clustering of the expression data showed that the MT and LM tissues had closely related expression profiles when compared to para-tumor normal tissue or primary CRC tissue from CRC patients without liver metastasis (NM), suggesting clonal and genetic similarity for these pairs (Fig. 3a). We identified a total of 2136 genes significantly changed in LM (Fig. 3b) when compared to PN or NM groups, in which 462 genes (21.6%) were enriched in metabolism pathways (P = 1.58E-17) (Fig. 3c). Among them, 256 (55.41%) of which were down-regulated and 206 (44.59%) of which were up-regulated.
Moreover, analysis from TCGA sequencing dataset identified a total of 6585 significantly changed genes in CLM (Fig. 3d). A total of 5632 genes were significantly changed (3788 down-regulated and 1844 up-regulated) in the MT group compared to the PN group; and 1709 genes were significantly changed (1697down-regulated and 12 up-regulated) in the MT group compared to the CRC tumor samples without liver metastasis (NM) group. Among that, 1254 genes were in common with DEmRNAs identified in our study (58.7%).

mRNA versus protein abundance in CLM

When compared with the 1386 DEproteins identified by LC-MS/MS, 362 DEmRNAs showed significant positive mRNA-protein correlation (Fig. 3e, left and middle panels). To determine whether the concordance between protein and mRNA variation is related to the biological function of the gene product, we performed KEGG enrichment analysis, which indicated that among the 362 significantly deregulated genes/proteins, 48 are enriched in metabolic pathways (P = 4.59E-5) (Fig. 3e, right panel). These findings further verify the role of metabolic pathway genes in CLM.

Impact of copy number alterations in CLM

We further performed global copy number variation (CNV) analysis to identify likely gene targets of focal alterations and to explore the impact of CNVs on mRNA and protein abundance and the potential correlation with LM. PN samples displayed scarcely any gains or losses, however, relative to the PN group, the LM and MT groups had 321 regions of significant focal amplification and 209 regions of significant focal deletion (Fig. 4a, b). In addition to several previously well-defined arm-level changes associated with carcinogenesis of CRC17, gains of 2q, 5p, 6p, 10q, 11p and 16p/q and deleted 18p/q were identified to contain the mRNA abundance variation (Fig. 4a, b). When compared with the correlation of the protein-CNV correlation (Spearman’s correlation coefficient 0.41; P < 0.05), the correlation between protein level and mRNA expression was much stronger (Spearman’s correlation coefficient 0.53; P < 0.01) (Fig. 4c). These results suggest that the mRNA transcript abundance is a relatively reliable predictor of protein abundance differences, but that copy number alterations showed little consistency with the protein level.
To further examine the potential role of CNV, we calculated the number of genes/proteins that also had CNV alteration. When compared with the 362 significantly changed genes/proteins, 112 were found with changed copy-number (Fig. 4d). Among those, chromosomes 1, 6, 7, 8, 12, 16, 19 and X contained the strongly hot spots driving global mRNA abundance variation (Fig. 4d), which highlights the importance of these regions in CLM.

Evaluation the prognostic and biological power of significantly dysregulated proteins in CLM

We next evaluated the clinical significance of 286 CRC patients from TCGA database for the 112 CNV-mRNA-protein correlated molecules. Our results showed that 4 up-regulated genes (HSP90AB1, COL1A2, FABP5 and BGN), which located in CNV hotspots (located in 6p21.1, 7q21.3, 8q21.13 and Xq28, respectively) were associated with prognosis of CRC patients (Fig. 5a). Kaplan-Meier survival analysis confirmed that high expression of HSP90AB1, COL1A2, FABP5 or BGN was significantly associated with a shorter overall survival (P < 0.05) (Fig. 5b). Among those, high expression of COL1A2 and BGN was extremely significantly associated with a shorter overall survival (P < 0.01) (Fig. 5b). Moreover, high expression of COL1A2 or BGN was positively associated with disease-free survival (P < 0.05) as determined by Kaplan-Meier survival analysis.
To investigate the biological role of these CNV-mRNA-protein correlated genes, which associated with the prognosis in CRC progression and liver metastasis, we established the CRC cell line SW480 to stably overexpress COL1A2 or BGN to perform the gain-of-function studies in vitro (Fig. 5c). We then tested the effect of cell migration by COL1A2 or BGN overexpression via wound-healing assay and observed significant improvement of cell motility by COL1A2 or BGN (P < 0.01) (Fig. 5d). By two-chamber transwell assays, we also showed that forced expression of COL1A2 or BGN markedly enhanced the transwell invasiveness of SW480 (P < 0.01) (Fig. 5e).

Somatic coding mutations in primary and metastatic CRC

To provide a comprehensive understanding of genetic abnormalities occurring in CLM, we used massively parallel paired-end sequencing technology to perform whole-exome solution-based hybrid capture sequencing of 2 triplet sample sets. The mean sequencing depth in the target regions was 80.28× (range 71.5 to 92.85). Analysis of the whole-exome sequencing data identified 27,778.5 mean point mutations (range 26,323 to 29,126). There were a variety of types of mutations identified, with T > A transversion being the most common nucleotide substitution (Fig. 6a). The distribution of CLM-related SNVs is shown in Fig. 6b. After filter analysis and exclusion of synonymous mutations, the numbers of indels and non-synonymous SNVs were calculated (Fig. 6c). In addition to some previously reported mutations, such as those in TP53, APC, KRAS, and PIK3CA [5], we identified 97 MT and LM-shared point mutations and 701 point mutations only existed in MT (Fig. 6d).
We further assessed the somatic gene mutations in an extended validation group of 44 paired normal colorectal tissues and CRC tissues by Sanger sequencing and nucleotide polymorphism genotype analysis. Subsequently, 175 nonsynonymous mutations within 171 genes were further verified.
In addition to the expected APC, TP53, SMAD4, PIK3CA and KRAS mutations, we found some new mutations that have not been reported to be involved in CLM including the TLL2A302S mutation, which was identified in both the MT and LM groups (Fig. 6e), and the KLF11D19N mutation, which was specific for the LM cohort (Fig. 6f). Moreover, our single nucleotide polymorphism genotype analysis revealed that 2 sites (FABP5A2T and HSP90AB1E299N) were frequent mutated only in the MT cohort, with mutation rates of 4.55% (2/44) (Fig. 6g) and 15.9% (7/44) (Fig. 6h), which suggest their potential roles in CLM.

Single amino acid variants (SAAVs) in CRC

A fundamental goal of proteogenomics is to identify protein coding alterations that are expressed at the protein level. However, standard database search approaches cannot identify variant peptides from MS/MS data. Therefore, we created a customized mutation database to search for SAAVs in CRC. A SAAV library was prepared using 113,844 mutated sites in CRC tissues from cBioport and our whole exome sequencing data, and 16,581 mutated proteins were identified, which constitute 82.08% of 20,201 proteins in the CRC standard protein library (Fig. 7a).
We determined the total numbers of mutated and non-mutated peptides and tumor-specific mutant peptides and found that mutated peptide numbers in MT samples were significantly increased (Fig. 7b), which indicates that the mutated peptide number has potential predictive value for CRC liver metastasis. Among those, 12 SAAVs in 8 proteins occurred only in NM patients (Fig. 7c; Table 2); and 13 proteins in 18 MT patients had 26 SAAVs; of which, 26 SAAVs in 6 proteins occurred in both NM and MT samples (Fig. 7d; Table 3), and 11 SAAVs in 5 proteins only occurred in MT samples (Fig. 7e; Table 4).
Table 2
SAAVs in NM-specifical sample
Protein name
Accession number
Wild Type peptide
Mutant peptide
Site of peptide
Site of SAAV
No. of SAAVs
RAB2A
P61019
IQEGVFDIDNEANGIK
IQEGVFDINNEANGIK
P61019_171_186
D179N
4
CKAP4
Q07065
ITIQAITEK
IAIQAITEK
Q07065_347_355
T348A
2
VIM
P08670
IIEEMIQR
IQEEMIQR
P08670_189_196
I190E
1
PABPC1
P11940
GFGFVCFSSPEDATK
GFGFVCFSSPEEATK
P11940_334_348
D345E
1
UQCRC1
P31930
ICTSVTESEVAR
ICTSATESEVAR
P31930_379_390
V383A
1
HNRNPM
P52272
INDIISNAIK
INEIISNAIK
P52272_372_381
D374E
1
ACTG2
P63267
CEEETTAPVCDNGSGICK
CEEETTAIVCDNGSGICK
P63267_2_19
P9I
1
LRG1
P02750
NAITGIPSGIFQASATIDTIVIK
NAITGIPPGIFQASATIDTIVIK
P02750_126_148
S133P
1
Table 3
SAAVs in NM & MT-share sample
Protein name
Accession number
Wild Type peptide
Mutant peptide
Site of peptide
Site of SAAV
No. of SAAVs
MYH9
P35579
AGVIAHIEEER
AGVITHIEEER
P35579_765_775
A769T
8
HSPA9
P38646
EQQIVIQSSGGISKDDIENMVK
EQQIVIQSSGGISNDDIENMVK
P38646_542_563
K555 N
8
HSP90AB1
P08238
NPDDITQEEYGEFYK
NPDDITQDEYGEFYK
P08238_292_306
E299N
3
ATP2A2
P16615
DIVPGDIVEIAVGDK
DIVPGDNVEIAVGDK
P16615_144_158
I150N
3
FABP5
Q01469
ATVQQIEGR
TTVQQIEGR
Q01469_2_10
A2T
2
XPO1
O14980
NVDIIKDPETVK
NVDIIQDPETVK
O14980_675_686
K680Q
2
Table 4
SAAVs in MT-specifical sample
Protein name
Accession number
Wild Type peptide
Mutant peptide
Site of peptide
Site of SAAV
No. of SAAVs
CCT6A
P40227
NAIDDGCVVPGAGAVEVAMAEAIIK
NAIDDGCVVPGAGAVEVAMAEAINK
P40227_400_424
I423N
4
CAT
P04040
NISVEDAAR
NISVEDVAR
P04040_244_252
A250V
2
ACTN1
P12814
VGWEQIITTIAR
VGWEQIITTITR
P12814_715_726
A725T
1
JUP
P14923
TMQNTSDIDTAR
TMQNTNDIDTAR
P14923_192_203
S197 N
1
ARF4
P18085
HYFQNTQGIIFVVDSNDR
HYFQNTQGIIFVVDSDDR
P18085_80_97
N95D
1
FAM3D
Q96BQ1
AFDMYSGDVMHIVK
SFDMYSGDVMHIVK
Q96BQ1_118_131
A118S
1
HLA-B
P01889
FISVGYVDDTQFVR
FIAVGYVDDTQFVR
P01889_46_59
S48A
1
To further evaluate the potential role of SAAVs, we examined the expression levels of the proteins with SAAVs. The expression of 6 NM & MT-shared (Fig. 8a) and 8 MT-specific mutated proteins was upregulated in CRC. The sites of the most frequently mutated three proteins, MYH9A769T, HSPA9K555N and CCT6AI423N, are shown (Fig. 8c). Furthermore, high MYH9 and CCT6A expression were each associated with shorter overall survival and disease-free survival (P < 0.05; Fig. 8d), which indicates that they have potential predictive values for CRC liver metastasis.

Discussion

CRC is the third most common malignancy and the second leading cause of cancer deaths in many countries. It develops from a benign adenomatous polyp into an invasive cancer, and nearly 50% of CRC patients develop CLM [17]. Without treatment, patients with colorectal hepatic metastases have a median survival of only 5–10 months, with less than 0.5% surviving beyond 5 years [18].
The molecular pathogenesis of CRC is associated with a variety of genetic changes that lead to the aberrant activation of proto-oncogenes and inactivation of tumor suppressor genes [19]. Characterization of CRC genomes has been elaborated by large-scale next-generation sequencing, which has yielded important insights into the genes and mechanisms that contribute to cancer development and progression. A handful of recurrently mutated genes, including APC, KRAS, TP53, and SMAD4, have been discovered by this method [20]. According to the classical tumor progression model of sporadic CRC proposed by Fearon and Vogelstein, APC mutation is involved in adenoma formation, followed by KRAS oncogenic mutation that promotes the transition from intermediate adenomas to carcinomas, with TP53 inactivation as a late event [21]. Subsequently, mutations in individual genes (including SMAD4) facilitate CRC metastasis [22]. Leveraging the next generation sequencing technology, TCGA Network has reported the common occurrence of mutations in additional genes, such as ARID1A, SOX9 and FAM123B, which also demonstrate that CRC is a highly genetically heterogeneous disease at the population level [5].
Understanding the genetic differences between primary colon cancer and their metastases to the liver is essential for devising a better therapeutic approach for this disease [23]. Therefore, research efforts have shifted from identifying driving mutations of carcinogenesis to genetic abnormalities during CRC progression in order to provide valuable insights into the clonal relationship and genetic differences between primary CRCs and matched colorectal liver metastasis [24]. A recent study reported high genomic concordance between primary colorectal carcinoma and metastases, which indicate that somatic mutations may accumulate within the microenvironment of a primary cancer before disseminating to their metastatic sites [25]. Consistent with this hypothesis, in this study, we employed primary CRC tumor samples from patients with liver metastasis to trace progressive disease and combined CNV, mRNA and protein profiling data to identify potentially relevant genes in amplified chromosomal regions. Our results revealed the importance of chromosomes X, 7, 16 and 1, which contain the four strongest hot spots driving global mRNA abundance variation. These results also provided new insights into the potential roles of PFDN4 and COL1A2 in CLM. We also created a customized mutation database of CRC to identify SAAVs that occur during CRC metastases to the liver. The results indicate that the mutated peptide number has potential prognosis value, which can be broadly extended to understand roles of SAAVs in other cancers.

Conclusions

To the best of our knowledge, this is the first comprehensive study to use proteogenomic profiling of primary CRCs from patients with or without liver metastasis to define the dominant events of metastatic lesions in terms of their expression and mutation. Our comprehensive integrative analysis of 44 colorectal tumor and normal pairs provides a number of insights into the biology of CLM and identifies potential therapeutic targets. Moreover, our characterization of the annotated metastatic CRC proteome clarifies the power of integrating genomics (SNVs) and proteomics (SAAVs). This approach provides new insights into the roles of these protein alterations in CLM, which can be broadly extended to understand the roles of protein mutation in other cancers.

Acknowledgments

We would like to thank Prof. Yongguo Yu and Yiping Shen for data analysis and critical discussion of the manuscript.

Funding

This study was supported partly by grants from the National Natural Science Foundation of China (81772932, 81472202, 81201535, 81302065 and 81702243), The Fundamental Research Funds for the Central Universities (22120170212 and 22120170117), Shanghai Natural Science Foundation (12ZR1436000), Program of Shanghai Academic/Technology Research Leader (18XD1403000), and Shanghai Municipal Commission of Health and Family Planning (201540228).

Availability of data and materials

The datasets supporting the conclusions of this article are included within the article.

Accession codes

Whole-exome sequencing data from this study are available for download through the NCBI Sequence Read Archive under accession number PRJNA358865. All RNA-sequencing (GSE92914) data have been deposited in the Gene Expression Omnibus. All of the MS proteomics data have been deposited to iProX (http://​www.​iprox.​org/​index) and can be accessed with the accession IPX00083203 and IPX00083210.
The study was examined and approved by the Ethics Committee of the Shanghai Tenth People’s Hospital, Tongji University School of Medicine (SHSY-IEC-PAP-16-24). Each participant provided their written informed consent to participate in this study.
Not applicable.

Competing interests

The authors declare that they have no competing interest.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://​creativecommons.​org/​licenses/​by/​4.​0/​), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated.
Literatur
1.
Zurück zum Zitat Torre LA, Bray F, Siegel RL, Ferlay J, Lortet-Tieulent J, Jemal A. Global cancer statistics, 2012. CA Cancer J Clin. 2015;65:87–108.CrossRef Torre LA, Bray F, Siegel RL, Ferlay J, Lortet-Tieulent J, Jemal A. Global cancer statistics, 2012. CA Cancer J Clin. 2015;65:87–108.CrossRef
2.
Zurück zum Zitat Miller KD, Siegel RL, Lin CC, Mariotto AB, Kramer JL, Rowland JH, et al. Cancer treatment and survivorship statistics, 2016. CA Cancer J Clin. 2016;66:271–89.CrossRef Miller KD, Siegel RL, Lin CC, Mariotto AB, Kramer JL, Rowland JH, et al. Cancer treatment and survivorship statistics, 2016. CA Cancer J Clin. 2016;66:271–89.CrossRef
3.
Zurück zum Zitat Kandoth C, McLellan MD, Vandin F, Ye K, Niu B, Lu C, et al. Mutational landscape and significance across 12 major cancer types. Nature. 2013;513:333–9.CrossRef Kandoth C, McLellan MD, Vandin F, Ye K, Niu B, Lu C, et al. Mutational landscape and significance across 12 major cancer types. Nature. 2013;513:333–9.CrossRef
4.
Zurück zum Zitat Liu Y, Zhang X, Han C, Wan G, Huang X, Ivan C, et al. TP53 loss creates therapeutic vulnerability in colorectal cancer. Nature. 2015;520:697–701.CrossRef Liu Y, Zhang X, Han C, Wan G, Huang X, Ivan C, et al. TP53 loss creates therapeutic vulnerability in colorectal cancer. Nature. 2015;520:697–701.CrossRef
5.
Zurück zum Zitat Network TCGA. Comprehensive molecular characterization of human colon and rectal cancer. Nature. 2012;487:330–7.CrossRef Network TCGA. Comprehensive molecular characterization of human colon and rectal cancer. Nature. 2012;487:330–7.CrossRef
6.
Zurück zum Zitat Zhang B, Wang J, Wang X, Zhu J, Liu Q, Shi Z, et al. Proteogenomic characterization of human colon and rectal cancer. Nature. 2014;513:382–7.CrossRef Zhang B, Wang J, Wang X, Zhu J, Liu Q, Shi Z, et al. Proteogenomic characterization of human colon and rectal cancer. Nature. 2014;513:382–7.CrossRef
7.
Zurück zum Zitat Neilson KA, Ali N, Muralidharan S, Mirzaei M, Mariani M, Assadourian G, et al. less label, more free: approaches in label-free quantitative mass spectrometry. Proteomics. 2011;11:535–53.CrossRef Neilson KA, Ali N, Muralidharan S, Mirzaei M, Mariani M, Assadourian G, et al. less label, more free: approaches in label-free quantitative mass spectrometry. Proteomics. 2011;11:535–53.CrossRef
8.
Zurück zum Zitat Bouckaert RR, Drummond AJ. bModelTest: phylogenetic model averaging. Mol Biol Evol. 2003;20:1978–85.CrossRef Bouckaert RR, Drummond AJ. bModelTest: phylogenetic model averaging. Mol Biol Evol. 2003;20:1978–85.CrossRef
9.
Zurück zum Zitat Polisetty RV, Gautam P, Sharma R, Harsha HC, Nair SC, Gupta MK, et al. LC-MS/MS analysis of differentially expressed glioblastoma membrane proteome reveals altered calcium signaling and other protein groups of regulatory functions. Mol Cell Proteomics. 2012;11:M111.013565.CrossRef Polisetty RV, Gautam P, Sharma R, Harsha HC, Nair SC, Gupta MK, et al. LC-MS/MS analysis of differentially expressed glioblastoma membrane proteome reveals altered calcium signaling and other protein groups of regulatory functions. Mol Cell Proteomics. 2012;11:M111.013565.CrossRef
10.
Zurück zum Zitat Luber CA, Cox J, Lauterbach H, Fancke B, Selbach M, Tschopp J, et al. Quantitative proteomics reveals subset-specific viral recognition in dendritic cells. Immunity. 2010;32:279–89.CrossRef Luber CA, Cox J, Lauterbach H, Fancke B, Selbach M, Tschopp J, et al. Quantitative proteomics reveals subset-specific viral recognition in dendritic cells. Immunity. 2010;32:279–89.CrossRef
11.
Zurück zum Zitat Forero-Castro M, Robledo C, Benito R, Abáigar M, África Martín A, Arefi M, et al. Genome-wide DNA copy number analysis of acute lymphoblastic leukemia identifies new genetic markers associated with clinical outcome. PLoS One. 2016;11:e0148972.CrossRef Forero-Castro M, Robledo C, Benito R, Abáigar M, África Martín A, Arefi M, et al. Genome-wide DNA copy number analysis of acute lymphoblastic leukemia identifies new genetic markers associated with clinical outcome. PLoS One. 2016;11:e0148972.CrossRef
12.
Zurück zum Zitat Abuín JM, Pichel JC, Pena TF, Amigo J. BigBWA: approaching the burrows-wheeler aligner to big data technologies. Bioinformatics. 2015;31:4003–5.PubMed Abuín JM, Pichel JC, Pena TF, Amigo J. BigBWA: approaching the burrows-wheeler aligner to big data technologies. Bioinformatics. 2015;31:4003–5.PubMed
13.
Zurück zum Zitat Cibulskis K, McKenna A, Fennell T, Banks E, DePristo M, Getz G. ContEst: estimating cross-contamination of human samples in next-generation sequencing data. Genome Res. 2010;20:1297–303.CrossRef Cibulskis K, McKenna A, Fennell T, Banks E, DePristo M, Getz G. ContEst: estimating cross-contamination of human samples in next-generation sequencing data. Genome Res. 2010;20:1297–303.CrossRef
14.
Zurück zum Zitat Li H, Durbin R. PAnnBuilder: an R package for assembling proteomic annotation data. Bioinformatics. 2009;25:1094–5.CrossRef Li H, Durbin R. PAnnBuilder: an R package for assembling proteomic annotation data. Bioinformatics. 2009;25:1094–5.CrossRef
15.
Zurück zum Zitat Wang K, Li M, Hakonarson H. Exome sequencing identifies MAX mutations as a cause of hereditary pheochromocytoma. Nucleic Acids Res. 2010;38:e164.CrossRef Wang K, Li M, Hakonarson H. Exome sequencing identifies MAX mutations as a cause of hereditary pheochromocytoma. Nucleic Acids Res. 2010;38:e164.CrossRef
16.
Zurück zum Zitat Tindall EA, Speight G, Petersen DC, Padilla EJ, Hayes VM. Novel Plexor SNP genotyping technology: comparisons with TaqMan and homogenous MassEXTEND MALDI-TOF mass spectrometry. Hum Mutat. 2007;28:922–7.CrossRef Tindall EA, Speight G, Petersen DC, Padilla EJ, Hayes VM. Novel Plexor SNP genotyping technology: comparisons with TaqMan and homogenous MassEXTEND MALDI-TOF mass spectrometry. Hum Mutat. 2007;28:922–7.CrossRef
17.
Zurück zum Zitat Ongen H, Andersen CL, Bramsen JB, Oster B, Rasmussen MH, Ferreira PG, et al. Putative cis-regulatory drivers in colorectal cancer. Nature. 2014;512:87–90.CrossRef Ongen H, Andersen CL, Bramsen JB, Oster B, Rasmussen MH, Ferreira PG, et al. Putative cis-regulatory drivers in colorectal cancer. Nature. 2014;512:87–90.CrossRef
18.
Zurück zum Zitat Oh BY, Hong HK, Lee WY, Cho YB. Animal models of colorectal cancer with liver metastasis. Cancer Lett. 2017;387(114–20.CrossRef Oh BY, Hong HK, Lee WY, Cho YB. Animal models of colorectal cancer with liver metastasis. Cancer Lett. 2017;387(114–20.CrossRef
19.
Zurück zum Zitat Calon A, Lonardo E, Berenguer-Llergo A, Espinet E, Hernando-Momblona X, Iglesias M, et al. Stromal gene expression defines poor-prognosis subtypes in colorectal cancer. Nat Genet. 2015;47:320–9.CrossRef Calon A, Lonardo E, Berenguer-Llergo A, Espinet E, Hernando-Momblona X, Iglesias M, et al. Stromal gene expression defines poor-prognosis subtypes in colorectal cancer. Nat Genet. 2015;47:320–9.CrossRef
20.
Zurück zum Zitat Sayagués JM, Corchete LA, Gutiérrez ML, Sarasquete ME, Del M, Bengoechea O, et al. Genomic characterization of liver metastases from colorectal cancer patients. Oncotarget. 2016;7:72908–22.CrossRef Sayagués JM, Corchete LA, Gutiérrez ML, Sarasquete ME, Del M, Bengoechea O, et al. Genomic characterization of liver metastases from colorectal cancer patients. Oncotarget. 2016;7:72908–22.CrossRef
21.
Zurück zum Zitat Zhou M, Yang H, Learned RM, Tian H, Ling L. Non-cell-autonomous activation of IL-6/STAT3 signaling mediates FGF19-driven hepatocarcinogenesis. Nat Commun. 2017;8:15433.CrossRef Zhou M, Yang H, Learned RM, Tian H, Ling L. Non-cell-autonomous activation of IL-6/STAT3 signaling mediates FGF19-driven hepatocarcinogenesis. Nat Commun. 2017;8:15433.CrossRef
22.
Zurück zum Zitat Cheng D, Zhao S, Tang H, Zhang D, Sun H, Yu F, et al. MicroRNA-20a-5p promotes colorectal cancer invasion and metastasis by downregulating Smad4. Oncotarget. 2016;7:45199–213.PubMedPubMedCentral Cheng D, Zhao S, Tang H, Zhang D, Sun H, Yu F, et al. MicroRNA-20a-5p promotes colorectal cancer invasion and metastasis by downregulating Smad4. Oncotarget. 2016;7:45199–213.PubMedPubMedCentral
23.
Zurück zum Zitat Chen LG, Xia YJ, Cui Y. Upregulation of miR-101 enhances the cytotoxic effect of anticancer drugs through inhibition of colon cancer cell proliferation. Oncol Rep. 2017;38:100–8.CrossRef Chen LG, Xia YJ, Cui Y. Upregulation of miR-101 enhances the cytotoxic effect of anticancer drugs through inhibition of colon cancer cell proliferation. Oncol Rep. 2017;38:100–8.CrossRef
24.
Zurück zum Zitat Han D, Wang M, Ma N, Xu Y, Jiang Y, Gao X. Long noncoding RNAs: novel players in colorectal cancer. Cancer Lett. 2015;361:13–21.CrossRef Han D, Wang M, Ma N, Xu Y, Jiang Y, Gao X. Long noncoding RNAs: novel players in colorectal cancer. Cancer Lett. 2015;361:13–21.CrossRef
25.
Zurück zum Zitat Ouzounova M, Lee E, Piranlioglu R, El Andaloussi A, Kolhe R, Demirci MF, et al. Monocytic and granulocytic myeloid derived suppressor cells differentially regulate spatiotemporal tumour plasticity during metastatic cascade. Nat Commun. 2017;8:14979.CrossRef Ouzounova M, Lee E, Piranlioglu R, El Andaloussi A, Kolhe R, Demirci MF, et al. Monocytic and granulocytic myeloid derived suppressor cells differentially regulate spatiotemporal tumour plasticity during metastatic cascade. Nat Commun. 2017;8:14979.CrossRef
Metadaten
Titel
Proteogenomic characterization and comprehensive integrative genomic analysis of human colorectal cancer liver metastasis
verfasst von
Yu-Shui Ma
Tao Huang
Xiao-Ming Zhong
Hong-Wei Zhang
Xian-Ling Cong
Hong Xu
Gai-Xia Lu
Fei Yu
Shao-Bo Xue
Zhong-Wei Lv
Da Fu
Publikationsdatum
01.12.2018
Verlag
BioMed Central
Erschienen in
Molecular Cancer / Ausgabe 1/2018
Elektronische ISSN: 1476-4598
DOI
https://doi.org/10.1186/s12943-018-0890-1

Weitere Artikel der Ausgabe 1/2018

Molecular Cancer 1/2018 Zur Ausgabe

Adjuvante Immuntherapie verlängert Leben bei RCC

25.04.2024 Nierenkarzinom Nachrichten

Nun gibt es auch Resultate zum Gesamtüberleben: Eine adjuvante Pembrolizumab-Therapie konnte in einer Phase-3-Studie das Leben von Menschen mit Nierenzellkarzinom deutlich verlängern. Die Sterberate war im Vergleich zu Placebo um 38% geringer.

Alectinib verbessert krankheitsfreies Überleben bei ALK-positivem NSCLC

25.04.2024 NSCLC Nachrichten

Das Risiko für Rezidiv oder Tod von Patienten und Patientinnen mit reseziertem ALK-positivem NSCLC ist unter einer adjuvanten Therapie mit dem Tyrosinkinase-Inhibitor Alectinib signifikant geringer als unter platinbasierter Chemotherapie.

Bei Senioren mit Prostatakarzinom auf Anämie achten!

24.04.2024 DGIM 2024 Nachrichten

Patienten, die zur Behandlung ihres Prostatakarzinoms eine Androgendeprivationstherapie erhalten, entwickeln nicht selten eine Anämie. Wer ältere Patienten internistisch mitbetreut, sollte auf diese Nebenwirkung achten.

ICI-Therapie in der Schwangerschaft wird gut toleriert

Müssen sich Schwangere einer Krebstherapie unterziehen, rufen Immuncheckpointinhibitoren offenbar nicht mehr unerwünschte Wirkungen hervor als andere Mittel gegen Krebs.

Update Onkologie

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