Skip to main content
Erschienen in: BMC Cancer 1/2019

Open Access 01.12.2019 | Research article

A germline mutation in Rab43 gene identified from a cancer family predisposes to a hereditary liver-colon cancer syndrome

verfasst von: Yanting Jiang, Yue Sun, Jiandong Hu, Nan Yu, Hui Liu, Jiankun Fan, Xuelian Ning, Yilan Li, Baogang Liu, Yihua Sun, Jinwei Zhang, Xiaohong Qiu, Songbin Fu, Chunshui Zhou, Hui Xu

Erschienen in: BMC Cancer | Ausgabe 1/2019

Abstract

Background

Hereditary cancer syndromes have inherited germline mutations which predispose to benign and malignant tumors. Understanding of the molecular causes in hereditary cancer syndromes has advanced cancer treatment and prevention. However, the causal genes of many hereditary cancer syndromes remain unknown due to their rare frequency of mutation.

Methods

A large Chinese family with a history of hereditary liver-colon cancer syndrome was studied. The genomic DNA was extracted from the blood samples of involved family members, whole-exome sequencing was performed to identify genetic variants. Functional validation of a candidate variant was carried out using gene expression, gene knockout and immunohistochemistry.

Results

The whole-exome of the proband diagnosed with colon cancer was sequenced in comparison with his mother. A total of 13 SNVs and 16 InDels were identified. Among these variants, we focused on a mutation of Rab43 gene, a GTPase family member involving in protein trafficking, for further validation. Sanger DNA sequencing confirmed a mutation (c: 128810106C > T, p: A158T) occurred in one allele of Rab43 gene from the proband, that heterozygous mutation also was verified in the genome of the proband’s deceased father with liver cancer, but not in his healthy mother and sister. Ectopic expression of the Rab43 A158T mutant in Huh7 cells led to more enhanced cell growth, proliferation and migration compared to the expression of wild type Rab43. Conversely, knockout of Rab43 in HepG2 cells resulted in slow cell growth and multiple nuclei formation and impaired activation of Akt. Finally, a positive correlation between the expression levels of Rab43 protein and cancer development in that family was confirmed.

Conclusions

A germline mutation of Rab43 gene is identified to be associated with the onset of a familial liver-colon cancer syndrome. Our finding points to a potential role of protein trafficking in the tumorigenesis of the familial cancer syndrome, and helps the genetic counseling to the affected family members.
Hinweise

Electronic supplementary material

The online version of this article (https://​doi.​org/​10.​1186/​s12885-019-5845-4) contains supplementary material, which is available to authorized users.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Abkürzungen
A158T
Alanine 158 Threonine
Crispr
Clustered regularly interspaced short palindromic repeats
ER
Endoplasmic reticulum
G12 V
Glycine 12 Valine
IHC
Immunohistochemistry
Indels
Insertions and deletions
MTT
3-(4,5)-dimethylthiazol (− 2-y1)-2,5-di- phenyltetrazolium bromide
SNVs
Single nucleotide variants

Background

Hereditary cancer syndromes refer to a group of cancer patients harboring germline mutations and these mutations predispose carriers to several benign and malignant tumors. So far, only a small number of hereditary cancer syndromes are extensively studied. For example, mutations in the TP53 gene are the cause of about 70% of Li-Fraumeni cancer syndrome [1]. Mutations in BRCA1 (17q21) and BRCA2 (13q12.3) have been linked to hereditary breast and ovarian cancer syndrome [2, 3]. Familial Adenomatous Polyposis (FAP) is caused by mutations in the adenomatous polyposis coli (APC) gene [4]. Germline mutations in DNA mismatch repair genes including MLH1, MSH2, MSH6, or PMS2 give rise to Lynch syndrome [5]. The understanding of molecular aberrations in hereditary cancer syndromes has dramatically advanced targeted cancer therapies in recent years. The knowledge gained from genetics of hereditary cancer syndromes can also help genetic counseling and cancer prevention [6]. However, despite a few of well-defined hereditary cancer syndromes described above, the causal mutations of the majority hereditary cancer syndromes are largely unclear due to their extremely low frequency of mutation. Here, we identified a large Chinese cancer family with a clear hereditary family history of liver and colon cancer. We employed whole-exome sequencing technology to analyze the genomic DNA from the blood of the proband in comparison with his mother and discovered a heterozygous germline mutation of Rab43 gene is associated with the onset of this familial liver and colon cancer syndrome.

Methods

Patients and DNA samples

The family with a history of hereditary cancers consisted of two persons with colorectal cancer and four persons with liver cancer spanning in three consecutive generations. Blood samples were collected from available family members except the fourth generation members because of their young ages and other concerns. For the proband’s deceased father diagnosed with liver cancer, DNA and liver tissue were obtained from the pathological archives of a local hospital where the patient was treated. All living participants involved in this research signed written informed consent forms. This research was approved by the Harbin Medical University ethics committee in accordance with Helsinki Declaration.

Genomic DNA preparation and whole-exome sequencing

Genomic DNA was extracted from peripheral leukocytes by DNA purification Kit (Qiagen) according to the manufactrue’s instructions. DNA concentrations, DNA purity and integrity were assessed by spectrophotometry (Nanodrop) and agarose gel electrophoresis and met the DNA sequencing requirements. The gDNA library was prepared using Ion Xpress Plus Fragment Library Kit (Thermo Fisher Scientific). Exome enrichment was performed by SureSelect™ target enrichment system (Illumina) in accordance with the manufacturer’s instructions. The enriched DNA samples were sequenced via paired 126 bp sequencing using a Hiseq2500 Sequencing System (Illumina).

Whole-exome sequencing data filtering analysis

A binary sequence alignment map (BAM) file was generated by comparison of the paired DNA samples. Somatic SNVs and indels were detected by comparison of the paired BAM files. The sequence reads were aligned to the human reference genome (UCSChg19) using the Burrows-Wheeler Aligner (BWA) with default parameters. Variants were identified using the Genome Analysis Toolkit (GATK). SNVs and indels were analyzed using Haplotype Caller and data were filtered by variant quality score recalibration. Relevant mutations in all of the genes were subsequently prioritized manually.

Variant validation, allele segregation analysis

Exome sequencing results for prioritized variants were validated using specific primers for amplification by polymerase chain reaction and Sanger sequencing. Segregation analysis of the prioritized variants was performed in additional family members (including a deceased member with hepatocellular cancer) whenever genomic DNA was available.

Immunohistochemistry (IHC)

IHC was carried out on paraffin-embedded sections of colon, liver cancer and their adjacent non-cancerous tissues obtained through surgery to investigate the expression levels of endogenous Rab43 with primary antibodies against human Rab43. A total of fifteen cancerous and fifteen adjacent non-cancerous tissue sections (slides) were analyzed for each liver or colon cancer patient. Each slide was blinded read under light microscopy by two board-certified pathologists. Ten randomly picked view fields from each slide were assessed. The staining intensity and proportion of positively stained cells were evaluated as described previously with minor modifications [7]. The intensity of Rab43 staining (brown color) was scored as follows: 0, negative; 1, weak; 2, moderate; 3, strong; And the percentage of positively stained cells was scored as follows: 0, < 5%; 1, 5–25%; 2, 26–50%; 3, 51–75%; 4, > 75%. The sum of each intensity score multiplied by its corresponding positive percentage score was regarded as the Rab43 staining score for the viewed field and assigned as follows: - for (0); + for (1–2); ++ for (3–5); +++ for (6–8); ++++ for (9–12).

Cell lines and tissue culture

The human cancer cell lines used in our research including HeLa (CCL-2), A549 (CCL-185), Skov-3 (HTB-77) and HepG2 (HB-8065) were purchased from American Type Culture Collection (ATCC, VA, USA). Cell line Huh7 (BNCC337690) was purchased from the Cell Bank of the Chinese Academy of Sciences (Shanghai, China). These cell lines were free of mycoplasma contamination as tested by a PCR method prior to our research. Cells were maintained in Dulbecco’s Modified Eagle Medium (Gibco, NY, USA) supplemented with 10% fetal bovine serum (Gibco), 100 unit/ml penicillin and 0.1% (w/v) streptomycin (Gibco) in a 5% CO2 humidified incubator at 37 °C.

Plasmid construction and cell line generation

The human Crispr-Cas9 guide RNA was designed based on the coding gene of Rab43 (GenBank accession number: AY166852). The primer pair for gRNA was: Rab43-cri-F3: 5′-caccggctactaccgcagtgccaat-3′, Rab43-cri-R3: 5′- aaacattggcactgcgg tagtagcc-3′. The primer pair was annealed using the following parameters: denatured at 95 °C for 5 min and cooled down to room temperature. The annealed primer pair was cloned into the Lenti-Crispr-V2 vector (a gift from Dr. Feng Zhang of MIT, USA) through BsmI restriction sites. The gRNA cloning was verified by Sanger sequencing. HepG2 cells were transfected with the plasmid Lenti-Crispr-V2-Rab43–3 by lipofectamin-3000 according to the manufacturer’s instruction. The stable transfected cells were selected by puromycin, and the resulting cell clones with desired mutations in Rab43 gene were verified by Sanger sequencing and Western blotting.
Wild type Rab43 gene was amplified from a gift plasmid (from Dr. John Cox of University of Tennessee) containing the wild type ORF of Rab43 by using two following specific primers with XhoI and EcoRI restriction sites. Rab43-cpXhoI-F: 5′-aattctcgagatggcagggccgggccca-3′, Rab43-cpEcoRI-R: 5′-aattgaattctcagcacccgcagcc ccag-3′. Wild type Rab43 ORF was subcloned into pcDNA3.1 (−)-3 × Flag via XhoI and EcoRI sites. Site-directed mutagenesis using following primers with the desired mutations (F: 5′- ctatgacatcctgtgtaccattgagacgtctg-3′, R: 5′-cagacgtctcaatggtacacaggat gtcatag-3′) was performed for Rab43 A158T mutant generation. The Huh-7 cells transfected with pcDNA3.1-(−)-3 × Flag-Rab43 A158T were selected by G418. Following primer pair was used for generating Crispr resistance Rab43 ORF in pcDNA3.1-(−)-3 × Flag plasmid. Cri-resi-3F: 5′-agctactatagatcagctgcagccgccatccttg-3′, Cri-resi-3R: 5′-caaggatggcggctgcagctgatctatagtagct-3′.

Colony formation assay

Cells were seeded at a density of 500 cells/well in 6-well plates. After 2 weeks, the cells were fixed with 4 ml of 4% paraformaldehyde for 15 min, and stained with 0.2% crystal violet for 10 min. The numbers of visible colonies with more than 50 cells per colony were counted. Colony formation was calculated from three independent experiments.

Cell proliferation assay

Cells were seeded in 96-well plates at the density of 1000 cells/well and incubated for the next day. The cell proliferation assays (MTT) were performed on day 1, 3 and 5 after seeding. Experiments were carried out in triplicate.

In vitro cell migration assay

Cell migration assay was performed by using wound healing method as described previously [8]. Briefly, cells were seeded into a six-well plate at a density of 5 × 105/well and allowed to culture overnight. Cells were then treated with mitomycin C (final concentration at 50μg/ml, Sigma) to inhibit cell proliferation for 2 h before scratches were generated by applying a 200 μL pipette tip. Cells were washed twice with PBS buffer in order to remove all the dislodged cells. Photographs were taken at 0 h, 24 h and 48 h after wounding. Migration of the cells was measured by determining the wound healing areas of control cells versus Rab43 overexpressed Huh7 cells. Assays were performed in triplicate.

Western blotting

Cells were lysed in RIPA lysis buffer. The lysates were sonicated and centrifuged at 12,000 rpm and 4 °C for 10 min. Cell lysates were quantified using BCA protein assay kit (Pierce) and normalized for protein content by RIPA buffer. After separation on SDS-PAGE, the proteins were then transferred to PVDF membrane (Millipore). The membranes were incubated with the primary antibody (1:1000) probing Rab43 (Santa Cruz), Akt (Cell Signaling), p-Akt (Ser473) (Cell Signaling),, phos-pP70S6K (Cell Signaling) or GAPDH (Sigma) over night at 4 °C and incubated with the fluorescein conjugated secondary antibodies (1:5000) for 1 h at room temperature. The proteins were detected and photographed using Odyssey imaging system (Li-COR biosciences). GAPDH was used as loading control. The signal intensity was quantified using the Image J software (NIH).

Statistical analysis

For statistical evaluation, data are expressed as mean ± standard deviation (SD). Statistical differences for comparison of cell growth, colony formation and wound healing were determined by Student’s t test or one-way ANOVA test. The Rab43 expression levels in colon and liver tissues were examined by Mann–Whitney test. All analyses were performed using SPSS for Windows 17.0 software (SPSS Inc., Chicago, IL, USA). p < 0.05 was considered statistically significant.

Results

Identification of germline mutations in a Chinese hereditary cancer family

We identified a large Chinese family with four generations living in Northeast of China with a clear history of hereditary liver and colon cancers. As of the date when blood samples were collected, a total of two colon cancers and four liver cancers and one colon adenoma have been diagnosed spanning the first three consecutive generations (Fig. 1). Those diseases display an obvious autosomal dominant pattern of inheritance. Furthermore, there is no affected member in the fourth generation most likely due to their relative young ages.
We sequenced the whole-exome of the proband who was diagnosed with colon cancer at the age of 54. Meanwhile, his mother’s whole-exome sequencing was performed in parallel for comparison. In addition, we also compared our sequencing results with the hg19 genome database maintained by University of California Santa Cruz as a third party reference. We used six bioinformatics tools to select for deleterious mutations, including PhyloP (score > 0.85), SIFT (score < 0.05), PolyPhen (score > 0.85), GERP (score > 2), Mutation Taster (score > 0.5), and LRT (score > 0.9), and only those mutations with four or more positive deleterious predictions were further considered. Eventually, we obtained 13 SNVs from 13 genes (Table 1) and 16 Indels from 15 genes (Table 2). Among these candidate genes, based on available information including their known functions, potential protein interactions and predicted effects on protein structure, we focused on the variant of Rab43 for further analysis. There is a C > T transition occurred on the anti-sense strand of one Rab43 allele in the genome of the proband, this mutation leads to substitution of alanine by threonine at the 158 residue on Rab43 protein. The heterozygous mutation status of Rab43 gene in the proband was confirmed by Sanger sequencing (Fig. 2a). The segregation of the mutant allele was analyzed in additional family members except all members from the fourth generation of that family. While the patient’s healthy mother and two sisters are homozygous for wild type Rab43 alleles, the genome of his father who died from liver cancer indeed harbors one of the mutant allele of Rab43 (Fig. 2b-d). These data demonstrated the mutant allele of Rab43 is clearly co-segregated with the familial cancer syndrome, though the segregation information is not available in the fourth generation currently. In addition, we performed protein structure analysis by using PyMol software [9] and found there is no overall structural change with the threonine substitution of alanine at residue 158 on Rab43 protein (Fig. 2e-f). There are many known oncogenic mutations that would interfere with the functions rather than the overall structures of the affected proteins. One prominent example of such mutations is the oncogenic Ras G12 V mutant. A previous study reported the overall structures between the GDP bound form of the normal Ras protein and the GDP bound form of the Ras G12 V are identical, suggesting it is the side chain on Valine12 rather than the conformation changes would impair the intrinsic GTPase activity associated with Ras [10]. In the case of Rab43 A158T, we propose it is the hydroxyl group on Threonine 158 rather than any structural changes that would enhance the functions associated with Rab43 protein, hinting strongly the identified Rab43 variant might be oncogenic with a gain of function.
Table 1
SNVs identified from the 54 year old proband in comparison with his mother
Chr
Position
rsID
Gene
DNA change
AA change
1
151,090,537
rs2864699
GABPB2
G/C
Q/H
1
248,569,476
OR2T1
G/T
D/Y
3
128,810,106
rs970572
RAB43
C/T a
A/T
4
17,609,087
LAP3
C/T
H/Y
7
6,692,019
rs2250356
ZNF316
C/G
11
118,401,572
rs535760315
TTC36
T/C
C/R
12
110,206,393
rs824998
FAM222A
G/A
C/Y
15
102,359,116
rs1370063626
OR4F15
C/T
H/Y
16
66,776,341
rs8055189
DYNC1LI2
A/G
F/L
16
67,860,078
rs188707401
TSNAXIP1
C/T
R/C
16
87,466,766
rs7499131
ZCCHC14
T/G
K/Q
16
87,760,390
rs4843689
KLHDC4
T/C
Y/C
22
17,600,584
CECR6
G/T
D/E
Chr chromosome and its number, rsID the indel ID obtained from dbSNP135 database, AA change amino acid change
aThe C to T transition in Rab43 gene reflects the sequencing results of the DNA changes on the anti-sense strand of Rab43 gene
Table 2
InDels identified from the 54 year old proband in comparisonwith his mother
Chr
Position
rsID
Gene
DNA change
AA change
3
54,959,059
rs370607786
LRTM1
T/−
H > Fs
3
75,787,349–75,787,353
ZNF717
CCCTG/−
T > Fs
3
97,887,844
rs377659479
OR5H15
T/−
F > Fs
4
70,360,942
UGT2B4
T/−
N > Fs
8
10,480,295
rs201192645
RP1L1
−/G
P > Fs
11
4,566,460
rs77893620
OR52M1
T/−
F > Fs
14
38,679,763
SSTR1
−/GCTCT
T > delins TLX
17
62,855,003
LRRC37A3
−/A
19
41,018,845
SPTBN4
C/−
Q > Fs
20
168,625
rs11396059
DEFB128
−/T
H > Fs
20
1,592,151–1,592,152
SIRPB1
G/−
L > Fs
20
1,592,154
SIRPB1
−/AC
D > Fs
20
2,674,430
EBF4
T/−
20
50,701,795
ZFP64
G/−
P > Fs
21
11,029,597
rs60459764
BAGE2,BAGE3
C/−
22
43,926,809–43,926,813
EFCAB6
AGAGT/−
Y > Fs
Chr chromosome and its number, rsID the indel ID obtained from dbSNP135 database, AA change amino acid change

Rab43 A158T variant exhibits oncogenic activity in liver cancer cells

Rab43 is a Rab family related small GTPase and regulates the sorting of a subset of protein cargo through the medial Golgi [1113]. Similar to mutations occurred on oncogene Ras [14, 15], the Rab43 mutation identified in our study might be oncogenic. In order to determine the functional consequence of Rab43 A158T, we subcloned wild type Rab43 gene and generated Rab43 A158T mutant gene by site directed mutagenesis. We screened an array of human cancer cell lines and found the expression levels of Rab43 are varied dramatically (Fig. 3a). Ectopically expressed both wild type Rab43 and Rab43 A158T mutant proteins in liver cancer cell line Huh7 with a very low level of endogenous Rab43 protein enhanced cell growth and proliferation (Fig. 3b-c), stimulated the colony formation on cultured plates (Fig. 3d-e) and promoted the migration of the transfected Huh7 cells in vitro (Fig. 3f-g). More strikingly, compared to the effects of the expressed wild type Rab43, the expressed Rab43 A158T mutant protein tends to confer more malignant transformation capacity to the transfected Huh7 cells (Fig. 3c-g). Interestingly, overexpression of Rab43A158T in another liver cancer cell line HepG2 with a high level of endogenous Rab43 expression has minimal effects on the growth, proliferation, colony formation and migration of the transfected HepG2 cells, indicating Rab43 A158T variant can substitute the function of WT Rab43 once wild type Rab43 is insufficient or lost completely. Taken together, our results strongly support Rab43 A158T variant possesses oncogenic activity and plays a driving role in the onset of the studied cancer syndrome, consistent with the autosomal dominant pattern of inheritance observed in that family.

Rab43 is required for Akt activation and Akt signaling

Rab43 has been implicated in glioma development previously [16], but the underlying mechanism is unclear. To further elucidate the molecular mechanism of Rab43 variant mediated tumorigenesis, we eliminated the Rab43 expression in HepG2 cells by Crispr-Cas9 gene editing technology targeting the exon 2 of Rab43 gene (Fig. 4a) and established a HepG2 Rab43 knockout cell line with 3 different reading frame shift mutations in the coding region of Rab43 gene, which cause complete loss of endogenous Rab43 protein expression in the Crispr-Rab43 HepG2 cells (Fig. 4a-b, for detailed Sanger sequencing results see Additional file 1: Figure S1). To our surprise, the Rab43 knockout HepG2 cells are enlarged and flattened with slow growth (Fig. 4c-d). Some knockout cells exhibit multi-nuclei, indicating problems in cell dividing and cell cycle progression. These observations suggest certain level of Rab43 is required for normal cell growth and cell cycle progression. To further reveal the molecular mechanism underlying Rab43 mediated regulation, the Akt activation in control (V2 vector transfected) and Crispr-Rab43 HepG2 cells was examined under normal culture conditions. We found the basal level of Akt activation was barely detectable in Crispr-Rab43 cells without any stimulation (Fig. 4e), rendering the reasonable comparison of Akt activation between the two cell lines unfeasible. Thus, we switched to look at the activation of Akt following insulin stimulation. Despite the fact that little activated Akt was detected in the insulin stimulated Rab43 knockout cells, Akt activation was dramatically increased in wild type Rab43 control cells following insulin stimulation, whereas the levels of total Akt protein in both cell lines were almost equal (Fig. 4e-g). Moreover, a similar pattern of the phosphorylation of p70S6K, a downstream target of Akt signaling, was observed (Fig. 4f-g), suggesting Akt-mTOR-p70S6K signaling is impaired upon loss of Rab43. Since it is well known that Rab43 involves in ER-Golgi trafficking, our data point to a potential role of protein trafficking in cell transformation and tumorigenesis.

High levels of Rab43 protein are correlated with the familial cancer syndrome

To further confirm the relevance of identified Rab43 variant to the onset of the studied familial cancer syndrome, we performed immunohistochemistry in colon tissues of the 54Y proband with colon cancer and in liver tissues of his farther died from liver cancer. We found high levels of Rab43 protein occurred in both colon and liver cancer tissues, whereas much less of Rab43 was detected in their adjacent non-cancerous tissues (Fig. 5a-c). Eventually, a total of 15 cancerous or non-cancerous tissue sections (slides) from the proband or his father were analyzed, the overall expression difference of Rab43 protein in between cancer tissues and non-cancerous tissues is statistically significant (Fig. 5b-d) (p < 0.001). These results clearly demonstrated there is a positive correlation between Rab43 protein expression level and the development of the familial liver- colon cancer syndrome. Given the fact that the proband and his father were heterozygous carriers of the Rab43 mutant allele, we are not sure whether the high levels of Rab43 observed in cancer tissues is contributed by elevated expression of Rab43 A158T alone. Nevertheless, the positive correlation between Rab43 and cancer development in the family strongly hints Rab43 A158T mutant might be the driving force for the onset of that familial liver-colon cancer syndrome.

Discussion

To identify susceptible genes for hereditary diseases, especially, hereditary cancer syndromes, two classes of approaches including functional screening and DNA genetic analysis are often exploited [17, 18]. Functional screening consists of gain-of-function screens (e.g. human cDNA expression screens) and loss-of-function screens (e.g. antisense application, RNA interference and Crispr-Cas9 gene editing) [19, 20]. As to DNA genetic analysis, whole genome sequencing, whole-exome sequencing and genome wide genetic linkage analyses are employed mostly [2123]. A couple of advantages are offered by whole-exome sequencing. First of all, this approach is a well-developed technology targeting coding genes only, which reduces the workload of DNA sequencing and data analysis significantly. Secondly, whole-exome sequencing is able to identify 100% heterozygous mutations at the 30x sequencing data coverage [24]. Lastly, the whole-exome sequencing data is less complex and more reliable compared to that of whole genome sequencing. As such, we used whole-exome sequencing to search for potential susceptible gene candidates for that familial liver and colon cancer syndrome.
Despite the fact that a large amount of efforts have been put into the identification of causal genetic mutations susceptible to hereditary cancers, however, there are still many unknown rare high-penetrance mutations predisposing to these cancers to be discovered yet [25, 26]. The recently discovered germ-line mutations in POLE and POLD1 genes from individuals with multiple colorectal adenomas, carcinoma, or both are excellent examples in support of above assumption [27]. Several layers of evidence indicate the familial liver-colon cancer syndrome studied by us differs from canonical hereditary colorectal cancer (CRC) and Lynch syndrome. First, we did not find any germline mutations in both APC and MUTYH genes [4, 28], two major causal mutations for CRC. Secondly, we also failed to identify mutations from mismatch repair genes including MLH1, MSH2, MSH6 and PMS [29, 30], which are major susceptible genes for Lynch syndrome. Thirdly, our studied family was affected by two types of cancers. As of the date when blood samples were collected, four persons were diagnosed with liver cancer and two persons were diagnosed with colon cancer in that family. These two diseases spanned in three consecutive generations display an obvious autosomal dominant inheritance pattern. So far, no such diseases were discovered in the fourth generation of that family, most likely due to their young ages. Lastly, our functional analysis demonstrated the property of Rab43 variant is oncogenic, in sharp contrast to the loss-of-function nature of mutations occurred in APC, MUTYH and mismatch repair genes, etc.
How a germline mutation on Rab43 gene is susceptible to hereditary cancer remains unclearly currently. Rab43 is a member of small GTPase family including Ras oncogene. The known function of Rab43 is to facilitate the ER-Golgi transportation of newly synthesized proteins in coordination with Arf/Arl/Rab family members [11, 31]. Additionally, Rab43 has been implicated in the Shiga toxin trafficking from cellular membrane to Golgi and biogenesis of Golgi complex [13, 32]. We demonstrated Akt activation requires intact Rab43. Upon loss of Rab43, there is a little of activated Akt, whereas the total level of Akt protein remains unchanged (Fig. 4e-f), suggesting some of the Rab43 mediated functions might be required for normal activation of Akt. Furthermore, Rab43 A158T mutant would promote Rab43 functions and stimulate the activation of Akt, leading to enhanced cell proliferation and transformation. By contrast, loss of Rab43 protein would impair Rab43 involved processes and impede the Akt activation. In support of our working model of Rab43 regulating Akt activation, Akt has been reported to facilitate Rab5 mediated endocytosis [33] and is able to promote the phosphorylation of Rab43 in pancreatic duct carcinoma cells [34]. Towards this end, more vigorous investigations are in need in the future.
On the other hand, proteins involving in ER-Golgi trafficking have been implicated in tumorigenesis. High level expression of Rab43 was reported in Chinese glioma cancer patients and correlated with the disease progression and poor prognosis [16] and cell adhesion and cell migration are also affected by Rab43 and other small GTPases [35]. Moreover, the oncogenic nature of Rab43 A158T mutant demonstrated by our experiments is in perfect agreement with the autosomal dominant pattern of inheritance of this familial liver-colon cancer syndrome, indicating a driving role of the Rab43 mutation in the onset of that hereditary cancer syndrome.

Conclusions

In summary, by using whole-exome sequencing, we identified that a germline mutation occurred on Rab43 gene is associated with the predisposition to a Chinese familial liver and colon cancer syndrome, exploring this mutant allele will not only help better understand the potential role of protein trafficking in tumorigenesis of this familial cancer syndrome, but also facilitate the genetic counseling to the affected families and offer more feasible prevention strategies in the clinic.

Acknowledgements

We thank Professor John Cox of University of Tennessee Health Science Center for generously providing the Rab43 cloning plasmid.
The study has been approved by the ethics committee of Harbin Medical University in accordance with Helsinki Declaration. Written informed consent was obtained from all persons involved in this study or their legal representative.
Not applicable.

Competing interests

The authors declare that they have no competing interests.
Open Access This 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.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Literatur
1.
Zurück zum Zitat Guha T, Malkin D. Inherited TP53 mutations and the Li-Fraumeni syndrome. Cold Spring Harb Perspect Med. 2017;7:4.CrossRef Guha T, Malkin D. Inherited TP53 mutations and the Li-Fraumeni syndrome. Cold Spring Harb Perspect Med. 2017;7:4.CrossRef
2.
Zurück zum Zitat Kwong A, Shin VY, Ho JC, Kang E, Nakamura S, Teo SH, Lee AS, Sng JH, Ginsburg OM, Kurian AW, et al. Comprehensive spectrum of BRCA1 and BRCA2 deleterious mutations in breast cancer in Asian countries. J Med Genet. 2016;53(1):15–23.CrossRef Kwong A, Shin VY, Ho JC, Kang E, Nakamura S, Teo SH, Lee AS, Sng JH, Ginsburg OM, Kurian AW, et al. Comprehensive spectrum of BRCA1 and BRCA2 deleterious mutations in breast cancer in Asian countries. J Med Genet. 2016;53(1):15–23.CrossRef
3.
Zurück zum Zitat Suszynska M, Klonowska K, Jasinska AJ, Kozlowski P. Large-scale meta-analysis of mutations identified in panels of breast/ovarian cancer-related genes - providing evidence of cancer predisposition genes. Gynecol Oncol. 2019;153(2):452-62.CrossRef Suszynska M, Klonowska K, Jasinska AJ, Kozlowski P. Large-scale meta-analysis of mutations identified in panels of breast/ovarian cancer-related genes - providing evidence of cancer predisposition genes. Gynecol Oncol. 2019;153(2):452-62.CrossRef
4.
Zurück zum Zitat Jasperson KW, Tuohy TM, Neklason DW, Burt RW. Hereditary and familial colon cancer. Gastroenterology. 2010;138(6):2044–58.CrossRef Jasperson KW, Tuohy TM, Neklason DW, Burt RW. Hereditary and familial colon cancer. Gastroenterology. 2010;138(6):2044–58.CrossRef
5.
Zurück zum Zitat Ligtenberg MJ, Kuiper RP, Chan TL, Goossens M, Hebeda KM, Voorendt M, Lee TY, Bodmer D, Hoenselaar E, Hendriks-Cornelissen SJ, et al. Heritable somatic methylation and inactivation of MSH2 in families with lynch syndrome due to deletion of the 3′ exons of TACSTD1. Nat Genet. 2009;41(1):112–7.CrossRef Ligtenberg MJ, Kuiper RP, Chan TL, Goossens M, Hebeda KM, Voorendt M, Lee TY, Bodmer D, Hoenselaar E, Hendriks-Cornelissen SJ, et al. Heritable somatic methylation and inactivation of MSH2 in families with lynch syndrome due to deletion of the 3′ exons of TACSTD1. Nat Genet. 2009;41(1):112–7.CrossRef
6.
Zurück zum Zitat Esteban-Jurado C, Garre P, Vila M, Lozano JJ, Pristoupilova A, Beltran S, Abuli A, Munoz J, Balaguer F, Ocana T, et al. New genes emerging for colorectal cancer predisposition. World J Gastroenterol. 2014;20(8):1961–71.CrossRef Esteban-Jurado C, Garre P, Vila M, Lozano JJ, Pristoupilova A, Beltran S, Abuli A, Munoz J, Balaguer F, Ocana T, et al. New genes emerging for colorectal cancer predisposition. World J Gastroenterol. 2014;20(8):1961–71.CrossRef
7.
Zurück zum Zitat Eckstein M, Jung R, Weigelt K, Sikic D, Stohr R, Geppert C, Agaimy A, Lieb V, Hartmann A, Wullich B, et al. Piwi-like 1 and −2 protein expression levels are prognostic factors for muscle invasive urothelial bladder cancer patients. Sci Rep. 2018;8(1):17693.CrossRef Eckstein M, Jung R, Weigelt K, Sikic D, Stohr R, Geppert C, Agaimy A, Lieb V, Hartmann A, Wullich B, et al. Piwi-like 1 and −2 protein expression levels are prognostic factors for muscle invasive urothelial bladder cancer patients. Sci Rep. 2018;8(1):17693.CrossRef
8.
Zurück zum Zitat Xu H, Sun H, Zhang H, Liu J, Fan F, Li Y, Ning X, Sun Y, Dai S, Liu B, et al. An ShRNA based genetic screen identified Sesn2 as a potential tumor suppressor in lung Cancer via suppression of Akt-mTOR-p70S6K signaling. PLoS One. 2015;10(5):e0124033.CrossRef Xu H, Sun H, Zhang H, Liu J, Fan F, Li Y, Ning X, Sun Y, Dai S, Liu B, et al. An ShRNA based genetic screen identified Sesn2 as a potential tumor suppressor in lung Cancer via suppression of Akt-mTOR-p70S6K signaling. PLoS One. 2015;10(5):e0124033.CrossRef
9.
Zurück zum Zitat Barcena C, Quesada V, De Sandre-Giovannoli A, Puente DA, Fernandez-Toral J, Sigaudy S, Baban A, Levy N, Velasco G, Lopez-Otin C. Exome sequencing identifies a novel mutation in PIK3R1 as the cause of SHORT syndrome. BMC Med Genet. 2014;15:51.CrossRef Barcena C, Quesada V, De Sandre-Giovannoli A, Puente DA, Fernandez-Toral J, Sigaudy S, Baban A, Levy N, Velasco G, Lopez-Otin C. Exome sequencing identifies a novel mutation in PIK3R1 as the cause of SHORT syndrome. BMC Med Genet. 2014;15:51.CrossRef
10.
Zurück zum Zitat Prive GG, Milburn MV, Tong L, de Vos AM, Yamaizumi Z, Nishimura S, Kim SH. X-ray crystal structures of transforming p21 ras mutants suggest a transition-state stabilization mechanism for GTP hydrolysis. Proc Natl Acad Sci U S A. 1992;89(8):3649–53.CrossRef Prive GG, Milburn MV, Tong L, de Vos AM, Yamaizumi Z, Nishimura S, Kim SH. X-ray crystal structures of transforming p21 ras mutants suggest a transition-state stabilization mechanism for GTP hydrolysis. Proc Natl Acad Sci U S A. 1992;89(8):3649–53.CrossRef
11.
Zurück zum Zitat Cox JV, Kansal R, Whitt MA. Rab43 regulates the sorting of a subset of membrane protein cargo through the medial Golgi. Mol Biol Cell. 2016;27(11):1834–44.CrossRef Cox JV, Kansal R, Whitt MA. Rab43 regulates the sorting of a subset of membrane protein cargo through the medial Golgi. Mol Biol Cell. 2016;27(11):1834–44.CrossRef
12.
Zurück zum Zitat Li C, Wei Z, Fan Y, Huang W, Su Y, Li H, Dong Z, Fukuda M, Khater M, Wu G. The GTPase Rab43 controls the anterograde ER-Golgi trafficking and sorting of GPCRs. Cell Rep. 2017;21(4):1089–101.CrossRef Li C, Wei Z, Fan Y, Huang W, Su Y, Li H, Dong Z, Fukuda M, Khater M, Wu G. The GTPase Rab43 controls the anterograde ER-Golgi trafficking and sorting of GPCRs. Cell Rep. 2017;21(4):1089–101.CrossRef
13.
Zurück zum Zitat Haas AK, Yoshimura S, Stephens DJ, Preisinger C, Fuchs E, Barr FA. Analysis of GTPase-activating proteins: Rab1 and Rab43 are key Rabs required to maintain a functional Golgi complex in human cells. J Cell Sci. 2007;120(Pt 17:2997–3010.CrossRef Haas AK, Yoshimura S, Stephens DJ, Preisinger C, Fuchs E, Barr FA. Analysis of GTPase-activating proteins: Rab1 and Rab43 are key Rabs required to maintain a functional Golgi complex in human cells. J Cell Sci. 2007;120(Pt 17:2997–3010.CrossRef
14.
Zurück zum Zitat Simanshu DK, Nissley DV, McCormick F. RAS proteins and their regulators in human disease. Cell. 2017;170(1):17–33.CrossRef Simanshu DK, Nissley DV, McCormick F. RAS proteins and their regulators in human disease. Cell. 2017;170(1):17–33.CrossRef
15.
Zurück zum Zitat Hunter JC, Manandhar A, Carrasco MA, Gurbani D, Gondi S, Westover KD. Biochemical and structural analysis of common Cancer-associated KRAS mutations. Mol Cancer Res. 2015;13(9):1325–35.CrossRef Hunter JC, Manandhar A, Carrasco MA, Gurbani D, Gondi S, Westover KD. Biochemical and structural analysis of common Cancer-associated KRAS mutations. Mol Cancer Res. 2015;13(9):1325–35.CrossRef
16.
Zurück zum Zitat Han MZ, Huang B, Chen AJ, Zhang X, Xu R, Wang J, Li XG. High expression of RAB43 predicts poor prognosis and is associated with epithelial-mesenchymal transition in gliomas. Oncol Rep. 2017;37(2):903–12.CrossRef Han MZ, Huang B, Chen AJ, Zhang X, Xu R, Wang J, Li XG. High expression of RAB43 predicts poor prognosis and is associated with epithelial-mesenchymal transition in gliomas. Oncol Rep. 2017;37(2):903–12.CrossRef
17.
Zurück zum Zitat Wang T, Wei JJ, Sabatini DM, Lander ES. Genetic screens in human cells using the CRISPR-Cas9 system. Science. 2014;343(6166):80–4.CrossRef Wang T, Wei JJ, Sabatini DM, Lander ES. Genetic screens in human cells using the CRISPR-Cas9 system. Science. 2014;343(6166):80–4.CrossRef
18.
Zurück zum Zitat Wan D, Gong Y, Qin W, Zhang P, Li J, Wei L, Zhou X, Li H, Qiu X, Zhong F, et al. Large-scale cDNA transfection screening for genes related to cancer development and progression. Proc Natl Acad Sci U S A. 2004;101(44):15724–9.CrossRef Wan D, Gong Y, Qin W, Zhang P, Li J, Wei L, Zhou X, Li H, Qiu X, Zhong F, et al. Large-scale cDNA transfection screening for genes related to cancer development and progression. Proc Natl Acad Sci U S A. 2004;101(44):15724–9.CrossRef
19.
Zurück zum Zitat Shalem O, Sanjana NE, Hartenian E, Shi X, Scott DA, Mikkelson T, Heckl D, Ebert BL, Root DE, Doench JG, et al. Genome-scale CRISPR-Cas9 knockout screening in human cells. Science. 2014;343(6166):84–7.CrossRef Shalem O, Sanjana NE, Hartenian E, Shi X, Scott DA, Mikkelson T, Heckl D, Ebert BL, Root DE, Doench JG, et al. Genome-scale CRISPR-Cas9 knockout screening in human cells. Science. 2014;343(6166):84–7.CrossRef
20.
Zurück zum Zitat Khorashad JS, Eiring AM, Mason CC, Gantz KC, Bowler AD, Redwine HM, Yu F, Kraft IL, Pomicter AD, Reynolds KR, et al. shRNA library screening identifies nucleocytoplasmic transport as a mediator of BCR-ABL1 kinase-independent resistance. Blood. 2015;125(11):1772–81.CrossRef Khorashad JS, Eiring AM, Mason CC, Gantz KC, Bowler AD, Redwine HM, Yu F, Kraft IL, Pomicter AD, Reynolds KR, et al. shRNA library screening identifies nucleocytoplasmic transport as a mediator of BCR-ABL1 kinase-independent resistance. Blood. 2015;125(11):1772–81.CrossRef
21.
Zurück zum Zitat Picelli S, Vandrovcova J, Jones S, Djureinovic T, Skoglund J, Zhou XL, Velculescu VE, Vogelstein B, Lindblom A. Genome-wide linkage scan for colorectal cancer susceptibility genes supports linkage to chromosome 3q. BMC Cancer. 2008;8:87.CrossRef Picelli S, Vandrovcova J, Jones S, Djureinovic T, Skoglund J, Zhou XL, Velculescu VE, Vogelstein B, Lindblom A. Genome-wide linkage scan for colorectal cancer susceptibility genes supports linkage to chromosome 3q. BMC Cancer. 2008;8:87.CrossRef
22.
Zurück zum Zitat Wheeler DA, Srinivasan M, Egholm M, Shen Y, Chen L, McGuire A, He W, Chen YJ, Makhijani V, Roth GT, et al. The complete genome of an individual by massively parallel DNA sequencing. Nature. 2008;452(7189):872–6.CrossRef Wheeler DA, Srinivasan M, Egholm M, Shen Y, Chen L, McGuire A, He W, Chen YJ, Makhijani V, Roth GT, et al. The complete genome of an individual by massively parallel DNA sequencing. Nature. 2008;452(7189):872–6.CrossRef
23.
Zurück zum Zitat Study C, Houlston RS, Webb E, Broderick P, Pittman AM, Di Bernardo MC, Lubbe S, Chandler I, Vijayakrishnan J, Sullivan K, et al. Meta-analysis of genome-wide association data identifies four new susceptibility loci for colorectal cancer. Nat Genet. 2008;40(12):1426–35.CrossRef Study C, Houlston RS, Webb E, Broderick P, Pittman AM, Di Bernardo MC, Lubbe S, Chandler I, Vijayakrishnan J, Sullivan K, et al. Meta-analysis of genome-wide association data identifies four new susceptibility loci for colorectal cancer. Nat Genet. 2008;40(12):1426–35.CrossRef
24.
Zurück zum Zitat Suleiman SH, Koko ME, Nasir WH, Elfateh O, Elgizouli UK, Abdallah MO, Alfarouk KO, Hussain A, Faisal S, Ibrahim FM, et al. Exome sequencing of a colorectal cancer family reveals shared mutation pattern and predisposition circuitry along tumor pathways. Front Genet. 2015;6:288.CrossRef Suleiman SH, Koko ME, Nasir WH, Elfateh O, Elgizouli UK, Abdallah MO, Alfarouk KO, Hussain A, Faisal S, Ibrahim FM, et al. Exome sequencing of a colorectal cancer family reveals shared mutation pattern and predisposition circuitry along tumor pathways. Front Genet. 2015;6:288.CrossRef
25.
Zurück zum Zitat Smith CG, Naven M, Harris R, Colley J, West H, Li N, Liu Y, Adams R, Maughan TS, Nichols L, et al. Exome resequencing identifies potential tumor-suppressor genes that predispose to colorectal cancer. Hum Mutat. 2013;34(7):1026–34.CrossRef Smith CG, Naven M, Harris R, Colley J, West H, Li N, Liu Y, Adams R, Maughan TS, Nichols L, et al. Exome resequencing identifies potential tumor-suppressor genes that predispose to colorectal cancer. Hum Mutat. 2013;34(7):1026–34.CrossRef
26.
Zurück zum Zitat DeRycke MS, Gunawardena SR, Middha S, Asmann YW, Schaid DJ, McDonnell SK, Riska SM, Eckloff BW, Cunningham JM, Fridley BL, et al. Identification of novel variants in colorectal cancer families by high-throughput exome sequencing. Cancer Epidemiol Biomark Prev. 2013;22(7):1239–51.CrossRef DeRycke MS, Gunawardena SR, Middha S, Asmann YW, Schaid DJ, McDonnell SK, Riska SM, Eckloff BW, Cunningham JM, Fridley BL, et al. Identification of novel variants in colorectal cancer families by high-throughput exome sequencing. Cancer Epidemiol Biomark Prev. 2013;22(7):1239–51.CrossRef
27.
Zurück zum Zitat Palles C, Cazier JB, Howarth KM, Domingo E, Jones AM, Broderick P, Kemp Z, Spain SL, Guarino E, Salguero I, et al. Germline mutations affecting the proofreading domains of POLE and POLD1 predispose to colorectal adenomas and carcinomas. Nat Genet. 2013;45(2):136–44.CrossRef Palles C, Cazier JB, Howarth KM, Domingo E, Jones AM, Broderick P, Kemp Z, Spain SL, Guarino E, Salguero I, et al. Germline mutations affecting the proofreading domains of POLE and POLD1 predispose to colorectal adenomas and carcinomas. Nat Genet. 2013;45(2):136–44.CrossRef
28.
Zurück zum Zitat Pearlman R, Frankel WL, Swanson B, Zhao W, Yilmaz A, Miller K, Bacher J, Bigley C, Nelsen L, Goodfellow PJ, et al. Prevalence and Spectrum of germline Cancer susceptibility gene mutations among patients with early-onset colorectal Cancer. JAMA Oncol. 2017;3(4):464–71.CrossRef Pearlman R, Frankel WL, Swanson B, Zhao W, Yilmaz A, Miller K, Bacher J, Bigley C, Nelsen L, Goodfellow PJ, et al. Prevalence and Spectrum of germline Cancer susceptibility gene mutations among patients with early-onset colorectal Cancer. JAMA Oncol. 2017;3(4):464–71.CrossRef
29.
Zurück zum Zitat Suter CM, Martin DI, Ward RL. Germline epimutation of MLH1 in individuals with multiple cancers. Nat Genet. 2004;36(5):497–501.CrossRef Suter CM, Martin DI, Ward RL. Germline epimutation of MLH1 in individuals with multiple cancers. Nat Genet. 2004;36(5):497–501.CrossRef
30.
Zurück zum Zitat Nicolaides NC, Papadopoulos N, Liu B, Wei YF, Carter KC, Ruben SM, Rosen CA, Haseltine WA, Fleischmann RD, Fraser CM, et al. Mutations of two PMS homologues in hereditary nonpolyposis colon cancer. Nature. 1994;371(6492):75–80.CrossRef Nicolaides NC, Papadopoulos N, Liu B, Wei YF, Carter KC, Ruben SM, Rosen CA, Haseltine WA, Fleischmann RD, Fraser CM, et al. Mutations of two PMS homologues in hereditary nonpolyposis colon cancer. Nature. 1994;371(6492):75–80.CrossRef
31.
Zurück zum Zitat Dejgaard SY, Murshid A, Erman A, Kizilay O, Verbich D, Lodge R, Dejgaard K, Ly-Hartig TB, Pepperkok R, Simpson JC, et al. Rab18 and Rab43 have key roles in ER-Golgi trafficking. J Cell Sci. 2008;121(Pt 16:2768–81.CrossRef Dejgaard SY, Murshid A, Erman A, Kizilay O, Verbich D, Lodge R, Dejgaard K, Ly-Hartig TB, Pepperkok R, Simpson JC, et al. Rab18 and Rab43 have key roles in ER-Golgi trafficking. J Cell Sci. 2008;121(Pt 16:2768–81.CrossRef
32.
Zurück zum Zitat Fuchs E, Haas AK, Spooner RA, Yoshimura S, Lord JM, Barr FA. Specific Rab GTPase-activating proteins define the Shiga toxin and epidermal growth factor uptake pathways. J Cell Biol. 2007;177(6):1133–43.CrossRef Fuchs E, Haas AK, Spooner RA, Yoshimura S, Lord JM, Barr FA. Specific Rab GTPase-activating proteins define the Shiga toxin and epidermal growth factor uptake pathways. J Cell Biol. 2007;177(6):1133–43.CrossRef
33.
Zurück zum Zitat Goto-Silva L, McShane MP, Salinas S, Kalaidzidis Y, Schiavo G, Zerial M. Retrograde transport of Akt by a neuronal Rab5-APPL1 endosome. Sci Rep. 2019;9(1):2433.CrossRef Goto-Silva L, McShane MP, Salinas S, Kalaidzidis Y, Schiavo G, Zerial M. Retrograde transport of Akt by a neuronal Rab5-APPL1 endosome. Sci Rep. 2019;9(1):2433.CrossRef
34.
Zurück zum Zitat Rhee M, Lee SH, Kim JW, Ham DS, Park HS, Yang HK, Shin JY, Cho JH, Kim YB, Youn BS, et al. Preadipocyte factor 1 induces pancreatic ductal cell differentiation into insulin-producing cells. Sci Rep. 2016;6:23960.CrossRef Rhee M, Lee SH, Kim JW, Ham DS, Park HS, Yang HK, Shin JY, Cho JH, Kim YB, Youn BS, et al. Preadipocyte factor 1 induces pancreatic ductal cell differentiation into insulin-producing cells. Sci Rep. 2016;6:23960.CrossRef
35.
Zurück zum Zitat Palamidessi A, Frittoli E, Ducano N, Offenhauser N, Sigismund S, Kajiho H, Parazzoli D, Oldani A, Gobbi M, Serini G, et al. The GTPase-activating protein RN-tre controls focal adhesion turnover and cell migration. Curr Biol. 2013;23(23):2355–64.CrossRef Palamidessi A, Frittoli E, Ducano N, Offenhauser N, Sigismund S, Kajiho H, Parazzoli D, Oldani A, Gobbi M, Serini G, et al. The GTPase-activating protein RN-tre controls focal adhesion turnover and cell migration. Curr Biol. 2013;23(23):2355–64.CrossRef
Metadaten
Titel
A germline mutation in Rab43 gene identified from a cancer family predisposes to a hereditary liver-colon cancer syndrome
verfasst von
Yanting Jiang
Yue Sun
Jiandong Hu
Nan Yu
Hui Liu
Jiankun Fan
Xuelian Ning
Yilan Li
Baogang Liu
Yihua Sun
Jinwei Zhang
Xiaohong Qiu
Songbin Fu
Chunshui Zhou
Hui Xu
Publikationsdatum
01.12.2019
Verlag
BioMed Central
Erschienen in
BMC Cancer / Ausgabe 1/2019
Elektronische ISSN: 1471-2407
DOI
https://doi.org/10.1186/s12885-019-5845-4

Weitere Artikel der Ausgabe 1/2019

BMC Cancer 1/2019 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.