Skip to main content
Erschienen in: Journal of Experimental & Clinical Cancer Research 1/2019

Open Access 01.12.2019 | Research

Apatinib potentiates irradiation effect via suppressing PI3K/AKT signaling pathway in hepatocellular carcinoma

verfasst von: Junbin Liao, Huilin Jin, Shaoqiang Li, Lixia Xu, Zhenwei Peng, Guangyan Wei, Jianting Long, Yu Guo, Ming Kuang, Qi Zhou, Sui Peng

Erschienen in: Journal of Experimental & Clinical Cancer Research | Ausgabe 1/2019

Abstract

Background

Limited effective intervention for advanced hepatocellular carcinoma (HCC) is available. This study aimed to investigate the potential clinical utility of apatinib, a highly selective inhibitor of the vascular endothelial growth factor receptor-2 (VEGFR2) tyrosine kinase, as a radiosensitizer in the treatment of HCC.

Methods

Four human HCC cell lines SMMC-7721, MHCC-97H, HCCLM3 and Hep-3B were treated with apatinib, irradiation or combination treatment. Colony formation assay, flow cytometry and nuclear γ-H2AX foci immunofluorescence staining were performed to evaluate the efficacy of combination treatment. RNA sequencing was conducted to explore the potential mechanism. The impact of combination treatment on tumor growth was assessed by xenograft mice models.

Results

Colony formation assay revealed that apatinib enhanced the radiosensitivity of HCC cell lines. Apatinib suppressed repair of radiation-induced DNA double-strand breaks. Flow cytometry analysis showed that apatinib increased radiation-induced apoptosis. Apatinib radiosensitized HCC via suppression of radiation-induced PI3K/AKT pathway. Moreover, an in vivo study indicated apatinib combined with irradiation significantly decreased xenograft tumor growth.

Conclusions

Our results indicate that apatinib has therapeutic potential as a radiosensitizer in HCC, and PI3K/AKT signaling pathway plays a critical role in mediating radiosensitization of apatinib.
Hinweise
Junbin Liao, Huilin Jin and Shaoqiang Li contributed equally to this work.

Supplementary information

Supplementary information accompanies this paper at https://​doi.​org/​10.​1186/​s13046-019-1419-1.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Abkürzungen
CCK-8
Cell counting kit-8
CMC
Carboxymethyl cellulose
DMEM
Dulbecco’s modified Eagle’s media
DMSO
Dimethyl sulfoxide
DSBs
Double-strand breaks
FBS
Fetal bovine serum
HCC
Hepatocellular carcinoma
HRR
Homologous recombination repair
IHC
Immunohistochemistry
RFA
Radiofrequency ablation
RT
Radiotherapy
SBRT
Stereotactic body radiotherapy
SER
Sensitizing enhancement ratio
SIRT
Selective internal radiation therapy
TKI
Tyrosine kinase inhibitor
VEGFR2
Vascular endothelial growth factor receptor-2
γ-H2AX
Phosphorylated histone H2AX

Background

Hepatocellular carcinoma (HCC) is one of the most prevalent malignancies and the third leading cause of cancer-related deaths worldwide [1]. Surgical resection, liver transplantation, and radiofrequency ablation (RFA) have been successfully developed as radical therapeutics for early stage HCC patients with well-preserved liver function [2]. However, those curative treatments are not always applicable, limited by the extent of disease, patient co-morbidities, tumor location and source of organs and so on. Moreover, over half of HCC is diagnosed at an advanced stage [3], with few effective treatments available.
Stereotactic body radiotherapy (SBRT), a non-invasive treatment, has been introduced into the treatment of HCC, even for advanced cases, and its efficacy and safety have been well characterized by several studies. SBRT provides excellent local control for HCC smaller than 2 cm, and overall survival at 1 and 2 years after SBRT was 74.1 and 46.3%, respectively [4]. More importantly, SBRT is still an effective treatment for patients with locally advanced primary liver malignancies. Recently, Bujold, et al. [5] reported that after SBRT treatment, the 1-year survival rate and local control rate of 102 patients with advanced HCC reached 55 and 87%, respectively, with a low risk of serious toxicity. With advances in radiation oncology technology, radiotherapy (RT) has now been recommended by the National Comprehensive Cancer Network Guidelines as a locoregional treatment option for inoperable HCC. Though intra- and extrahepatic spreading are still the predominant failure patterns for SBRT [4], it is worthwhile to explore how to increase its treatment potency, considering the abovementioned advantages of SBRT. SBRT in combination with other local or systemic therapies has been considered to be effective tactics to augment therapeutic potency [6, 7].
Sorafenib, a multitargeted tyrosine kinase inhibitor (TKI) and the only FDA-approved first-line targeted drug for patients with advanced HCC currently, yields to extend overall survival by less than 3 months in 2 large phase III clinical trials [8, 9]. More importantly, increasing clinical evidences indicate that radiotherapy and tyrosine kinase inhibitor therapy have synergistic antitumor effect. Yoshiyuki et al. [10] showed radiotherapy in combination with sorafenib dramatically prolonged overall survival of patients with advanced HCC compared to sorafenib alone (31.2 months vs. 12.2 months). However, SBRT combined with sorafenib has a high incidence of severe toxic side effects [11], which is intolerable for patients. However, apatinib [12], a novel tyrosine kinase inhibitor, highly selectively inhibits vascular endothelial growth factor receptor-2 tyrosine kinase (VEGFR-2) activity and thereby suppresses tumor growth by inhibiting tumor angiogenesis. Some studies [1316] have revealed that apatinib shows encouraging antitumor activities and tolerable toxicities in several solid tumors, including HCC. Although well tolerated in patients with advanced HCC, apatinib monotherapy has limited control over HCC [17, 18], necessitating combinatorial administration of apatinib with other therapies for efficacy augmentation. Apatinib combined with chemotherapy or immunotherapy has achieved promising efficacy in several tumors [14, 15, 19, 20]. Several clinical studies [21, 22] have indicated favorable efficacy of the combined treatment of apatinib with radiotherapy with controllable and tolerable adverse reactions. However, whether radiotherapy combined with apatinib can bring clinical benefits to patients with HCC and its underlying mechanisms remain unknown.
Therefore, our study aimed to explore the effect of apatinib combined with radiotherapy on HCC through both in vitro and in vivo assays, investigate the potential mechanism, and provide a strong theoretical basis for exploration of clinical combinations of radiation and apatinib in HCC. In present study, we showed that apatinib improved the radiosensitivity of HCC cells by inhibiting PI3K/AKT pathway.

Materials and methods

Cell culture and Apatinib preparation

Human hepatocellular carcinoma cell lines SMMC-7721, MHCC-97H, HCCLM3, Hep-3B were purchased from the Cell Bank of the Chinese Academy of Sciences (Shanghai, China) and confirmed by STR. Cells were either maintained in RPMI 1640 or Dulbecco’s Modified Eagle’s Media (DMEM, Sigma, USA) supplemented with 10% fetal bovine serum (FBS, Gibco, NY, USA) and 1% penicillin–streptomycin.
Apatinib was obtained from Jiangsu Hengrui Medicine Co., Ltd. (Nanjing, China). Apatinib was dissolved in dimethyl sulfoxide (DMSO) for in vitro studies or in 0.5% (w/v) carboxymethyl cellulose (CMC) for in vivo experiment.

Cell irradiation

HCC cells pretreated with/without apatinib for 24 h at 37 °C were exposed to ionizing radiation using X-ray linear accelerator (RS2000, RadSource, Suwanee, USA) at a dose rate of 1.24 Gy/min.

Cell viability assay

Four HCC cells were seeded into 96-well plates at 4 × 103 cells/well, respectively. After 24 h, cells were exposed to apatinib at various concentrations in fresh complete medium for another 48 h. The cell viability was determined using the Cell Counting Kit-8 (CCK-8, Dojindo, Japan) according to the manufacturer’s protocol. Each group was set up in quadruplicate.

Colony formation assay

HCC cells were seeded into six-well plates and allowed to attach for 24 h. After 24 h pretreatment with DMSO or apatinib, supernatant was changed with fresh medium and cells were irradiated with different doses of X-ray radiation (2–8 Gy). Two to three weeks after irradiation, cells were fixed with 4% paraformaldehyde for 20 min followed by staining with 0.5% crystal violet at room temperature for 10 min. The colonies containing ≥50 cells per dish were counted. Cell survival curves were fitted with the multi-target, single-hit model. Sensitizing enhancement ratio (SER) was determined as the ratio of the mean inactivation dose under radiation-only conditions divided by the mean inactivation dose after apatinib plus irradiation treatment.

Cell cycle and apoptosis analysis

For cell cycle analysis, 12 h after irradiation, cells were collected and then fixed with 70% ice-cold ethanol at − 20 °C overnight. Before flow cytometric analysis, cells were washed with PBS and then cocultured with 0.25 mg/ml RNase A and 50 μg/ml propidium iodide (PI) for 30 min at 37 °C in the dark. Samples were analyzed by flow cytometry (CytoFlex, BD Biosciences, USA) and data were analysed using ModFit 5.1 software. For apoptosis analysis, cells were collected, washed with PBS, and stained with Annexin V-FITC and PI (Annexin V, FITC Apoptosis Detection Kit, Dojindo, Japan) for 15 min at 37 °C in the dark. Samples were analyzed by flow cytometry and data were analysed using FlowJo 10.0 software. The Annexin-V+/PI- cells were in the early phase of the apoptotic process; the Annexin-V+/PI+ cells indicated late apoptosis. The percentages of both groups of cells were computed. Each group was set up in triplicate.

Immunofluorescence

Approximately, 4 × 104 cells were seeded on coverslips in 24-well plates and pretreated with apatinib or DMSO for 24 h and then irradiated with 4 Gy X-ray. After fixed in 4% paraformaldehyde, cells were blocked with 5% goat serum, then incubated with anti-Phospho-Histone H2AX antibody overnight at 4 °C and Alexa Fluor488-conjugated anti-rabbit antibody (Invitrogen, USA) for detection. After stained with DAPI (Fluoroshied with DAPI, Sigma, USA), samples were photographed with immunofluorescence microscopy.

Western blots

Cell extracts were prepared on ice using cell lysis buffer (KeyGEN BioTECH, Nanjing, China) and phosphatase inhibitors (Sigma) as needed, boiled for 10 min under reducing conditions, and frozen at − 20 °C until use. 10–40 μg of protein were electrophoresed in 8% or 12% sodium dodecyl sulfate–polyacrylamide gel electrophoresis (SDS-PAGE) and blotted onto Immobilon PVDF membranes (Millipore). Signals were visualized by western blotting using primary antibodies (Additional file 1: Table S1), followed by corresponding peroxidase-conjugated secondary antibodies and Immobilion Western Chemilum HRP Substrate (Millipore, USA).

RNA-sequencing

RNA from SMMC-7721 and MHCC-97H cells treated with 4 Gy radiation alone or radiaton plus apatinib was isolated and enriched for mRNA with MicroPoly (A) Purist (Ambion, Austin, TX, USA). The quality and quantity of mRNA were evaluated with a Bioanalyzer 2100 (Agilent Technologies, Santa Clara, CA, USA), and 0.5–1 mg mRNA for each sample was used for RNA sequencing (Illumina HiSeq 2000 platform). The significance of digital gene expression profiles was used to identify differential expression genes between the two groups. The pathway enrichment analysis was performed in Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways using KOBAS software.

Immunohistochemistry and TUNEL assay

For IHC, deparaffinized tumor sections were treated with sodium citrate buffer (pH 6.0) for antigen retrieval at boiling temperature for 2.5 min, and then 3% hydrogen peroxide for 10 min to quenched endogenous peroxidase activity, blocked and incubated with primary antibodies at 4 °C overnight. The Vectastain Elite ABC Kit (Vector Laboratories, Inc. USA) and DAB kit (Dako, Denmark) were used to amplify and detect signals. In Situ Cell Death Detection Kit, Fluorescein (Roche, USA) was used for TUNEL assay to examine the apoptosis rate in the tumor tissues. The deparaffinized sections were pretreated with Proteinase K for 30 min, quenched endogenous fluorescent signal, and then incubated in TUNEL reaction mixture for 1 h at 37 °C. The nucleus was stained with DAPI. Apoptosis rate was calculated in 5 microscopic fields of each sample (four samples from each group).

In vivo study

Male BALB/c nude mice (3–4 weeks of age) were housed in specific pathogen-free facilities. The protocols involving animals were approved by the Institutional Ethics Committee for Clinical Research and Animal Trials of the First Affiliated Hospital, Sun Yat-sen University. SMMC-7721 (5 × 106) cells were inoculated into the right flanks of mice. After 7 days, mice were randomly divided into four groups (n = 8): (1) carboxymethyl cellulose (CMC) (negative control); (2) Apatinib; (3) CMC + irradiation; and (4) Apatinib + irradiation. CMC and apatinib were administered via oral gavage at doses of 150 mg/kg once a day. Tumors were irradiated with 7 Gy X-ray using RS2000 X-ray linear accelerator at a dose rate of 1.24 Gy/min on day 13 and 15. Tumor dimensions were measured every 3 days, and the tumor volume was calculated using V = length×width2/2. At day 34, mice were euthanized and tumor tissues were harvested for TUNEL and immunohistochemistry detection.

Statistical analysis

All means were calculated from at least three independent experiments, with error bars representing the SEM. Two-way ANAVO analysis of variance and unpaired t-test were performed using GraphPad Prism 5.0 (GraphPad Software, San Diego, USA). Group differences were considered statistically significant when p<0.05.

Results

Apatinib enhances radiosensitivity of HCC cells

To determine the effect of apatinib on growth of HCC cell lines, SMMC-7721, MHCC-97H, HCCLM3, Hep-3B were incubated with different concentrations of apatinib for 48 h, and then cell viability was detected by Cell Counting Kit-8 (CCK-8) assays. We observed that the cell viability was suppressed by apatinib in a dose-dependent manner (Fig. 1a). The IC50 was 23.8 μM, 17.2 μM, 20.9 μM and 14.8 μM in SMMC-7721, MHCC-97H, HCCLM3 and Hep-3B, respectively. Based on the cell viability, 15 μM apatinib was used for SMMC-7721 and 10 μM apatinib was selected for MHCC-97H, HCC-LM3, and Hep-3B for further experiments.
The inhibitory effect of apatinib in combination with irradiation on the growth of HCC cells was assessed by cell counting assay. SMMC-7721, MHCC-97H, HCCLM3, Hep-3B cells were pretreated with apatinib or DMSO for 24 h and then exposed to 4 Gy or sham radiation. As compared to irradiation alone, cell growth was remarkably suppressed in apatinib pretreatment plus X-ray group at different time points, especially at day 4 (Fig. 1b-e). To further confirm the radiosensitizing effects of apatinib, colony formation assay was performed. Colony formation revealed pretreatment with apatinib plus 4 Gy X-ray irradiation significantly reduced the survival rate in all four HCC cell lines, as compared with cells treated with irradiation alone. The sensitivity enhancement ratio (SER) was 1.22, 1.22, 1.19 and 1.24 in SMMC-7721, MHCC-97H, HCCLM3 and Hep-3B, respectively (Fig. 1f-i). These data indicated that apatinib sensitized HCC cell lines to irradiation in vitro.

Apatinib suppresses repair of radiation-induced DNA double-strand breaks

The major cellular impact of ionizing radiation is to induce DNA double-strand breaks (DSBs) and trigger the DNA damage repair response [23]. To determine whether apatinib pretreatment would enhance radiation-induced DNA damage and interfere with the DNA damage repair process, immunofluorescence was performed to evaluate foci of phosphorylated histone H2AX (γ-H2AX), which is a sensor of DNA strand breaks and promotes efficient DSBs repair [24]. We found that almost 100% HCC cells were γ-H2AX positive regardless of irradiation or combination treatment at an early time points (1 h post IR). In contrast, γ-H2AX positive cell remained notably more in apatinib pretreatment plus irradiation than that in irradiation alone at 24 h post IR (66.7% ± 3.8% compared to 45% ± 4.9% in SMMC-7721, and 61.7% ± 4.3% compared to 46.3% ± 4.1% in MHCC-97H, 58.7% ± 6.1% compared to 31% ± 4.7% in HCCLM3, 67.3% ± 2.7% compared to 54.7% ± 2.9% in Hep-3B, all p<0.05, Fig. 2a-b), which reflected inefficient repair of DSBs. In addition to immunofluorescence, we observed parallel increases in double-strand DNA breaks at 1 h and 24 h post IR as detected by Western blot (Fig. 2c). Together, these results suggested that apatinib reduced the DNA damage repair activity in HCC cell lines.

Apatinib increases radiation-induced apoptosis in HCC cells

Radiation-induced DNA damage triggers cell cycle G1 or G2 arrest, allowing time for cells to repair DNA damage. Apatinib has also been reported to induce cell cycle arrest at G1 or G2 arrest [25, 26]. To investigate whether the radiosensitization of apatinib was related to cell cycle redistribution, PI staining using flow cytometry was performed to assess the DNA content. 4 Gy IR alone induced a typical G2/M-phase arrest in all four HCC cell lines after 24 h, while apatinib increased the G0/G1 population. For SMMC-7721 cell, the radiation-induced G2/M-phase arrest was further enhanced by combination with apatinib, while such effect didn’t exist in other three cell lines (Additional file 2: Figure S1).
To further investigate the reason behind the growth inhibition induced by radiosensitization of apatinib in HCC cell lines, flow cytometry and Western blot were used to determine the level of apoptosis. Cell apoptosis was assessed quantitatively by flow cytometry after Annexin V-FITC and PI staining. The results showed that 4 Gy X-rays alone moderately increased the apoptotic population of SMMC-7721, MHCC-97H, HCCLM3, Hep-3B cells compared with sham-treatment cancer cells. Pretreatment with apatinib remarkably enhanced the radiation-induced apoptosis in SMMC-7721, MHCC-97H, HCCLM3, Hep-3B cells, as compared to the radiation group (14.41% ± 1.097, 15.63 ± 1.21, 15.25 ± 1.08 and 24.55% ± 3.441%, vs 7.87% ± 0.75, 8.5% ± 0.29, 8.08 ± 0.61, 10.14% ± 0.67%, respectively, all p<0.01) (Fig. 3a-d). Consistently, apatinib pretreatment further increased the level of cleaved caspase 3 and cleaved caspase 9, cleaved-PARP, which are key indicators of apoptosis (Fig. 3e). Taken together, these results demonstrated that apatinib enhanced radiation-induced cell apoptosis.

Apatinib blocks PI3K/AKT pathway to sensitize HCC cells to radiation

To explore the potential mechanism how apatinib regulates radiosensitivity, we performed RNA-sequencing in combined treatment group and radiation group using SMMC-7721 and MHCC-97H cells. As shown in Fig. 4a, pathway enrichment analysis revealed that PI3K-AKT signaling pathway was significantly affected in both SMMC-7721 and MHCC-97H cell lines. It is reported that PI3K/AKT cascade is up-regulated by radiation involved in mechanism of radiation resistance, and impaired PI3K/AKT signaling pathway can inhibit DNA double-strand break repair and improve radiosensitivity [27, 28]. We therefore speculated that apatinib might affect the radiosensitivity of HCC cell lines through inhibition of PI3K/AKT signaling pathway. Western blot analysis revealed that apatinib decreased the level of phosphorylated PI3K and phosphorylated AKT in both SMMC-7721 and MHCC-97H cell lines. Although PI3K/AKT cascade was up-regulated by radiation, apatinib pretreatment effectively inhibited activation of PI3K/AKT pathway (Fig. 4b). Moreover, RAD51, which plays a major role in homologous recombination repair (HRR) during DSBs repair [29], was decreased in apatinib plus irradiation group as compared to irradiation alone. Consistently, γ-H2AX still remained at a higher level in combined treatment compared to irradiation alone (Fig. 2c).
To further confirm whether the radiosensitization effect of apatinib was attributed to inhibition of PI3K/AKT pathway, LY294002, a highly selective inhibitor of PI3K, and MK2206, an allosteric inhibitor of AKT, were used for further study. The effect of PI3K or AKT inhibitors plus irradiation on apoptosis was assessed quantitatively by Annexin V-PI staining. Inhibition of PI3K plus irradiation markedly increased cell apoptosis as compared to irradiation alone in both SMMC-7721 and MHCC-97H  cell lines (Fig. 5a). Similarly, AKT inhibitor plus irradiation significantly enhanced cell apoptosis in both cell lines (Fig. 5b). Consistent with previous reports, irradiation upregulated the phosphorylated PI3K and phosphorylated AKT, while inhibition of PI3K diminished the irradiation-induced effects and decreased RAD51 level in both SMMC-7721 and MHCC-97H cell lines(Fig. 5c). Suppression of AKT with inhibitor showed similar effect of the phosphorylated AKT diminishment and RAD51 down-regulating (Fig. 5d). Taken together, these data indicated that apatinib sensitized HCC to radiation via suppression of PI3K/AKT pathway.

Apatinib enhances xenograft tumor growth delay in mice receiving radiotherapy

SMMC-7721 xenograft models were established to further verify the radiosensitization effect of apatinib on HCC. SMMC-7721 cells were subcutaneously inoculated into the right flanks of BABL/c nude mice. Mice were randomly divided into four groups when mice first developed a palpable mass (4 mm in diameter): Ctrl (n = 8), apatinib (n = 8), irradiation (n = 8), and irradiation with apatinib (n = 7). The diagram of therapy design was showed (Fig. 6a). As expected, tumor weight and tumor volumes were mildly decreased in apatinib or irradiation group (0.6556 g ± 0.1586 or 0.616 g ± 0.1849) compared to 1.823 g ± 0.2472 in ctrl group (p<0.01, p<0.01, respectively). The irradiation combined with apatinib group effectively suppressed the growth of SMMC-7721 xenograft tumor models compared to irradiation treatment alone (at day 34) (apatinib plus irradiation vs. irradiation alone: 0.14 g ± 0.07221 vs. 0.616 g ± 0.1849, p<0.05, Fig. 6b-d). Besides, the combination treatment was well tolerated in all animals as it did not significantly affect body weight gain patterns in mice (Fig. 6e). Moreover, compared to radiotherapy group, combination treatment group presented with significantly increased TUNEL staining of cell apoptosis and significantly reduced expression of proliferation marker Ki67 (Fig. 6f). These results indicated that apatinib acted as a radiosensitizer in HCC in vivo, which was consistent with the in vitro data described above.

Discussion

SBRT [5] and selective internal radiation therapy (SIRT) using yttrium-90 microspheres [30] have shown favorable therapeutic effects on HCC. In order to improve the efficacy and safety of radiotherapy, many radiosensitizers have been combined with radiotherapy [31]. Preclinical and clinical studies have shown that anti-angiogenic drugs, such as sorafenib and sunitinib [32], can increase the sensitivity of the radiotherapy effect on cancer. Apatinib, as a highly selective VEGFR2 inhibitor, has potential in the treatment of advanced HCC. However, there is no report to evaluate whether apatinib can enhance the therapeutic effect of radiotherapy on HCC. In our study, we found that radiotherapy combined with apatinib presented a synergistic anti-tumor effect through both in vitro and in vivo assays, which was mediated by induction of cell apoptosis and inhibition of PI3K/AKT pathway.
Previous studies have shown that the fraction size of 4–8 Gy and the total dose of 30–40 Gy are tolerable for patients with HCC, so 4 Gy, an effective and safe dose, was chosen for our in vitro study. We observed that the reproducing ability of HCC cells was dose-dependently inhibited by radiation as detected via colony formation assay (Fig. 1f-i). In combination with apatinib, the inhibition of reproduction ability caused by radiotherapy was more obvious, indicating that apatinib can sensitize HCC cells to radiotherapy (Fig. 1f-i). Increased apoptosis in cancer cells plays an important role in the efficacy of radiotherapy. In present study, apatinib combined with 4 Gy radiation significantly increased apoptosis as detected by Annexin V/PI and flow cytometry assay (Fig. 3a-d). Furthermore, the protein level of cleaved PARP, cleaved caspase-9 and cleaved caspase-3 was notably elevated in the combination group (Fig. 3e). Consistent with the in vitro results, apoptosis induced by radiation could be remarkably enhanced by the administration of apatinib in subcutaneous tumor model (Fig. 6f). Moreover, several studies reported that apatinib itself was capable of inducing tumor cell apoptosis, including anaplastic thyroid carcinoma [33], colorectal cancer [34], and osteosarcoma [25]. Therefore, our study demonstrated that apatinib may increase tumor radiation sensitivity by inducing cell apoptosis.
Radiation-related cell pro-survival signaling pathways include suppression of apoptosis induction, cell cycle arrest and promotion of DNA repair, all of which synergistically protect tumor cells from radiation damage ultimately [35]. Transcriptome sequencing was performed on two HCC cell lines treated with the combination therapy or the radiotherapy alone in present study. By using the pathway enrichment analysis, we identified PI3K/AKT signaling pathway was involved in both HCC cell lines (Fig. 4a). We observed that radiation could promote cancer cell survival through igniting the PI3K/AKT pathway, which, however, could be effectively inhibited by administration of apatinib. And, apatinib could downregulate RAD51 expression, which is a dysfunction marker of DNA repair (Fig. 4b). In fact, a considerable number of studies [3638] have demonstrated that inhibition of PI3K/AKT signaling pathway could enhance the radiosensitivity of tumor cells both in vitro and in vivo. In addition, increased sensitivity to radiation by inhibiting PI3K/AKT pathway is associated with a decreased DNA repair capacity.
Although many radiosensitizers have been studied in HCC, the clinical applications so far have not been satisfactory [39]. As previous studies reported, antiangiogenic drugs sensitized tumors to radiotherapy through normalization of tumor blood vessels [32, 40]. Tumor vessels are characterized as high vessel density, tortuosity, dilation, chaotic, and lack of pericytes coverage, which cause tumor microenvironment hypoxia [41, 42]. Radiation depends on oxygen presence to generate free radicals to kill tumor cells and inadequate oxygen perfusion may attenuate radiotherapy effect [43]. Antiangiogenic drugs could normalize tumor blood vessels, reduce vessel density and improve oxygen perfusion so as to augment radiation effect. This normalization effect exerted most at about 1 week’s optimal time window after drug administration [42]. Given the above evidences, in the present study, we designed our treatment schedule to perform irradiation therapy at day 6 and day 8 after the first delivery of apatinib in mice xenograft tumor model. We observed that tumor blood vessel density decreased significantly in irradiation plus apatinib treatment group as compared to monotherapy group or control group (Additional file 3: Figure S2).
In HCC treatment, sunitinib has been showed with more adverse events even with several treatment-related deaths [44]. When compared with sorafenib, not only was sunitinib significantly inferior to sorafenib, but also sunitinib had more adverse events and toxicity [45]. However, as a first line drug for advanced HCC, sorafenib, when combined with radiotherapy, was reported to cause some fatal side effect to several patients [11]. What’s more, several studies illustrated that radiation sensitization effect of sorafenib should be attributed to inhibition of VEGFR-2 [46, 47]. Apatinib is a highly selective VEGFR-2 tyrosine kinase inhibitors, which was confirmed as a safe and effective concurrent treatment in HCC [15]. In this aspect, apatinib may have advantage over sorafenib and sunitinib as a radiosensitizer in HCC. Thus, in present study, we investigated apatinib as a radiosensitizer in HCC and found synergistic antitumor effect of apatinib and radiotherapy on HCC through inhibiting PI3K-AKT signaling pathway.

Conclusions

In conclusion, the present study demonstrates that apatinib enhances the radiosensitivity of HCC through both in vitro and in vivo assays, and PI3K/AKT signaling pathway plays a critical role in mediating this effect. Our finding indicates the therapeutic potential of apatinib as a radiosensitizer for HCC.

Supplementary information

Supplementary information accompanies this paper at https://​doi.​org/​10.​1186/​s13046-019-1419-1.

Acknowledgements

Not applicable.
This research was approved by the Institutional Ethics Committee for Clinical Research and Animal Trials of the First Affiliated Hospital, Sun Yat-sen University.
All authors have agreed to publish this manuscript.

Competing interests

The authors declare that they have no competing interests.
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.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Literatur
1.
Zurück zum Zitat Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA, Jemal A. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2018;68:394–424.PubMed Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA, Jemal A. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2018;68:394–424.PubMed
2.
Zurück zum Zitat Bruix J, Reig M, Sherman M. Evidence-based diagnosis, staging, and treatment of patients with hepatocellular carcinoma. Gastroenterology. 2016;150:835–53.PubMedCrossRef Bruix J, Reig M, Sherman M. Evidence-based diagnosis, staging, and treatment of patients with hepatocellular carcinoma. Gastroenterology. 2016;150:835–53.PubMedCrossRef
3.
Zurück zum Zitat Llovet JM, Zucman-Rossi J, Pikarsky E, Sangro B, Schwartz M, Sherman M, Gores G. Hepatocellular carcinoma. Nat Rev Dis Primers. 2016;2:16018.PubMedCrossRef Llovet JM, Zucman-Rossi J, Pikarsky E, Sangro B, Schwartz M, Sherman M, Gores G. Hepatocellular carcinoma. Nat Rev Dis Primers. 2016;2:16018.PubMedCrossRef
4.
Zurück zum Zitat Wahl DR, Stenmark MH, Tao Y, Pollom EL, Caoili EM, Lawrence TS, et al. Outcomes after stereotactic body radiotherapy or radiofrequency ablation for hepatocellular carcinoma. J Clin Oncol. 2016;34:452–9.PubMedCrossRef Wahl DR, Stenmark MH, Tao Y, Pollom EL, Caoili EM, Lawrence TS, et al. Outcomes after stereotactic body radiotherapy or radiofrequency ablation for hepatocellular carcinoma. J Clin Oncol. 2016;34:452–9.PubMedCrossRef
5.
Zurück zum Zitat Bujold A, Massey CA, Kim JJ, Brierley J, Cho C, Wong RKS, et al. Sequential phase I and II trials of stereotactic body radiotherapy for locally advanced hepatocellular carcinoma. J Clin Oncol. 2013;31:1631–9.PubMedCrossRef Bujold A, Massey CA, Kim JJ, Brierley J, Cho C, Wong RKS, et al. Sequential phase I and II trials of stereotactic body radiotherapy for locally advanced hepatocellular carcinoma. J Clin Oncol. 2013;31:1631–9.PubMedCrossRef
6.
Zurück zum Zitat Barker HE, Paget JTE, Khan AA, Harrington KJ. The tumour microenvironment after radiotherapy: mechanisms of resistance and recurrence. Nat Rev Cancer. 2015;15:409–25.PubMedPubMedCentralCrossRef Barker HE, Paget JTE, Khan AA, Harrington KJ. The tumour microenvironment after radiotherapy: mechanisms of resistance and recurrence. Nat Rev Cancer. 2015;15:409–25.PubMedPubMedCentralCrossRef
8.
Zurück zum Zitat Llovet JM, Ricci S, Mazzaferro V, Hilgard P, Gane E, Blanc JF, et al. Sorafenib in advanced hepatocellular carcinoma. N Engl J Med. 2008;359:378–90.CrossRefPubMed Llovet JM, Ricci S, Mazzaferro V, Hilgard P, Gane E, Blanc JF, et al. Sorafenib in advanced hepatocellular carcinoma. N Engl J Med. 2008;359:378–90.CrossRefPubMed
9.
Zurück zum Zitat Cheng AL, Kang YK, Chen Z, Tsao CJ, Qin S, Kim JS, et al. Efficacy and safety of sorafenib in patients in the Asia-Pacific region with advanced hepatocellular carcinoma: a phase III randomised, double-blind, placebo-controlled trial. Lancet Oncol. 2009;10:25–34.PubMedCrossRef Cheng AL, Kang YK, Chen Z, Tsao CJ, Qin S, Kim JS, et al. Efficacy and safety of sorafenib in patients in the Asia-Pacific region with advanced hepatocellular carcinoma: a phase III randomised, double-blind, placebo-controlled trial. Lancet Oncol. 2009;10:25–34.PubMedCrossRef
10.
Zurück zum Zitat Wada Y, Takami Y, Matsushima H, Tateishi M, Ryu T, Yoshitomi M, et al. The safety and efficacy of combination therapy of sorafenib and radiotherapy for advanced hepatocellular carcinoma: a retrospective study. Intern Med. 2018;57:1345–53.PubMedCrossRef Wada Y, Takami Y, Matsushima H, Tateishi M, Ryu T, Yoshitomi M, et al. The safety and efficacy of combination therapy of sorafenib and radiotherapy for advanced hepatocellular carcinoma: a retrospective study. Intern Med. 2018;57:1345–53.PubMedCrossRef
11.
Zurück zum Zitat Chen S, Lin L, Kuo Y, Liang J, Kuo C, Chiou J. Phase 2 study of combined sorafenib and radiation therapy in patients with advanced hepatocellular carcinoma. Int J Radiat Oncol Biol Phys. 2014;88:1041–7.PubMedCrossRef Chen S, Lin L, Kuo Y, Liang J, Kuo C, Chiou J. Phase 2 study of combined sorafenib and radiation therapy in patients with advanced hepatocellular carcinoma. Int J Radiat Oncol Biol Phys. 2014;88:1041–7.PubMedCrossRef
12.
Zurück zum Zitat Tian S, Quan H, Xie C, Guo H, Lü F, Xu Y, et al. YN968D1 is a novel and selective inhibitor of vascular endothelial growth factor receptor-2 tyrosine kinase with potent activity in vitro and in vivo. Cancer Sci. 2011;102:1374–80.PubMedCrossRef Tian S, Quan H, Xie C, Guo H, Lü F, Xu Y, et al. YN968D1 is a novel and selective inhibitor of vascular endothelial growth factor receptor-2 tyrosine kinase with potent activity in vitro and in vivo. Cancer Sci. 2011;102:1374–80.PubMedCrossRef
13.
Zurück zum Zitat Li J, Qin S, Xu J, Xiong J, Wu C, Bai Y, et al. Randomized, double-blind, placebo-controlled phase III trial of apatinib in patients with chemotherapy-refractory advanced or metastatic adenocarcinoma of the stomach or gastroesophageal junction. J Clin Oncol. 2016;34:1448–54.PubMedCrossRef Li J, Qin S, Xu J, Xiong J, Wu C, Bai Y, et al. Randomized, double-blind, placebo-controlled phase III trial of apatinib in patients with chemotherapy-refractory advanced or metastatic adenocarcinoma of the stomach or gastroesophageal junction. J Clin Oncol. 2016;34:1448–54.PubMedCrossRef
14.
Zurück zum Zitat Lan CY, Wang Y, Xiong Y, Li JD, Shen JX, Li YF, et al. Apatinib combined with oral etoposide in patients with platinum-resistant or platinum-refractory ovarian cancer (AEROC): a phase 2, single-arm, prospective study. Lancet Oncol. 2018;19:1239–46.PubMedCrossRef Lan CY, Wang Y, Xiong Y, Li JD, Shen JX, Li YF, et al. Apatinib combined with oral etoposide in patients with platinum-resistant or platinum-refractory ovarian cancer (AEROC): a phase 2, single-arm, prospective study. Lancet Oncol. 2018;19:1239–46.PubMedCrossRef
15.
Zurück zum Zitat Xu JM, Zhang Y, Jia R, Yue CY, Chang L, Liu RR, et al. Anti-PD-1 antibody SHR-1210 combined with apatinib for advanced hepatocellular carcinoma, gastric or esophagogastric junction cancer: an open-label, dose escalation and expansion study. Clin Cancer Res. 2019;25:515–23.PubMedCrossRef Xu JM, Zhang Y, Jia R, Yue CY, Chang L, Liu RR, et al. Anti-PD-1 antibody SHR-1210 combined with apatinib for advanced hepatocellular carcinoma, gastric or esophagogastric junction cancer: an open-label, dose escalation and expansion study. Clin Cancer Res. 2019;25:515–23.PubMedCrossRef
16.
Zurück zum Zitat Hu X, Zhang J, Xu B, Jiang Z, Ragaz J, Tong Z, et al. Multicenter phase II study of apatinib, a novel VEGFR inhibitor in heavily pretreated patients with metastatic triple-negative breast cancer. Int J Cancer. 2014;135:1961–9.PubMedCrossRef Hu X, Zhang J, Xu B, Jiang Z, Ragaz J, Tong Z, et al. Multicenter phase II study of apatinib, a novel VEGFR inhibitor in heavily pretreated patients with metastatic triple-negative breast cancer. Int J Cancer. 2014;135:1961–9.PubMedCrossRef
17.
Zurück zum Zitat Zhen L, Jiali C, Yong F, Han X, Hongming P, Weidong H. The efficacy and safety of apatinib treatment for patients with unresectable or relapsed liver cancer: a retrospective study. J Cancer. 2018;9:2773–7.PubMedPubMedCentralCrossRef Zhen L, Jiali C, Yong F, Han X, Hongming P, Weidong H. The efficacy and safety of apatinib treatment for patients with unresectable or relapsed liver cancer: a retrospective study. J Cancer. 2018;9:2773–7.PubMedPubMedCentralCrossRef
18.
Zurück zum Zitat Yu WC, Zhang KZ, Chen SG, Liu WF. Efficacy and safety of apatinib in patients with intermediate/advanced hepatocellular carcinoma: a prospective observation study. Medicine (Baltimore). 2018;97:e9704.CrossRef Yu WC, Zhang KZ, Chen SG, Liu WF. Efficacy and safety of apatinib in patients with intermediate/advanced hepatocellular carcinoma: a prospective observation study. Medicine (Baltimore). 2018;97:e9704.CrossRef
19.
Zurück zum Zitat Lu W, Jin XL, Yang C, Du P, Jiang FQ, Ma JP, et al. Comparison of efficacy between TACE combined with apatinib and TACE alone in the treatment of intermediate and advanced hepatocellular carcinoma: a single-center randomized controlled trial. Cancer Biol Ther. 2017;18:433–8.PubMedPubMedCentralCrossRef Lu W, Jin XL, Yang C, Du P, Jiang FQ, Ma JP, et al. Comparison of efficacy between TACE combined with apatinib and TACE alone in the treatment of intermediate and advanced hepatocellular carcinoma: a single-center randomized controlled trial. Cancer Biol Ther. 2017;18:433–8.PubMedPubMedCentralCrossRef
20.
Zurück zum Zitat Wei X, Liu L, Guo X, Wang Y, Zhao J, Zhou S. Light-activated ROS-responsive nanoplatform codelivering apatinib and doxorubicin for enhanced chemo-photodynamic therapy of multidrug-resistant tumors. ACS Appl Mater Interfaces. 2018;10:17672–84.PubMedCrossRef Wei X, Liu L, Guo X, Wang Y, Zhao J, Zhou S. Light-activated ROS-responsive nanoplatform codelivering apatinib and doxorubicin for enhanced chemo-photodynamic therapy of multidrug-resistant tumors. ACS Appl Mater Interfaces. 2018;10:17672–84.PubMedCrossRef
21.
Zurück zum Zitat Zhao F, Tian W, Zeng M, Xia J, Hu H, Hao X, et al. Apatinib alone or combined with radiotherapy in metastatic prostate cancer: results from a pilot, multicenter study. Oncotarget. 2017;8:110774–84.PubMedPubMedCentral Zhao F, Tian W, Zeng M, Xia J, Hu H, Hao X, et al. Apatinib alone or combined with radiotherapy in metastatic prostate cancer: results from a pilot, multicenter study. Oncotarget. 2017;8:110774–84.PubMedPubMedCentral
22.
Zurück zum Zitat Zhao C, Zhang Q, Qiao W. Significant efficacy and well safety of apatinib combined with radiotherapy in NSCLC: case report. Medicine (Baltimore). 2017;96:e9276.CrossRef Zhao C, Zhang Q, Qiao W. Significant efficacy and well safety of apatinib combined with radiotherapy in NSCLC: case report. Medicine (Baltimore). 2017;96:e9276.CrossRef
23.
Zurück zum Zitat Rogakou EP, Pilch DR, Orr AH, Ivanova VS, Bonner WM. DNA double-stranded breaks induce histone H2AX phosphorylation on serine 139. J Biol Chem. 1998;273:5858–68.PubMedCrossRef Rogakou EP, Pilch DR, Orr AH, Ivanova VS, Bonner WM. DNA double-stranded breaks induce histone H2AX phosphorylation on serine 139. J Biol Chem. 1998;273:5858–68.PubMedCrossRef
24.
Zurück zum Zitat Tsai CL, Liu WL, Hsu FM, Yang PS, Yen RF, Tzen KY, et al. Targeting histone deacetylase 4/ubiquitin-conjugating enzyme 9 impairs DNA repair for radiosensitization of hepatocellular carcinoma cells in mice. Hepatology. 2017;67:586–99.PubMedCrossRef Tsai CL, Liu WL, Hsu FM, Yang PS, Yen RF, Tzen KY, et al. Targeting histone deacetylase 4/ubiquitin-conjugating enzyme 9 impairs DNA repair for radiosensitization of hepatocellular carcinoma cells in mice. Hepatology. 2017;67:586–99.PubMedCrossRef
25.
Zurück zum Zitat Liu K, Ren T, Huang Y, Sun K, Bao X, Wang S, et al. Apatinib promotes autophagy and apoptosis through VEGFR2/STAT3/BCL-2 signaling in osteosarcoma. Cell Death Dis. 2017;8:e3015.PubMedPubMedCentralCrossRef Liu K, Ren T, Huang Y, Sun K, Bao X, Wang S, et al. Apatinib promotes autophagy and apoptosis through VEGFR2/STAT3/BCL-2 signaling in osteosarcoma. Cell Death Dis. 2017;8:e3015.PubMedPubMedCentralCrossRef
27.
Zurück zum Zitat Bussink J, van der Kogel AJ, Kaanders JH. Activation of the PI3-K/AKT pathway and implications for radioresistance mechanisms in head and neck cancer. Lancet Oncol. 2008;9:288–96.PubMedCrossRef Bussink J, van der Kogel AJ, Kaanders JH. Activation of the PI3-K/AKT pathway and implications for radioresistance mechanisms in head and neck cancer. Lancet Oncol. 2008;9:288–96.PubMedCrossRef
28.
Zurück zum Zitat Prevo R, Deutsch E, Sampson O, Diplexcito J, Cengel K, Harper J, et al. Class I PI3 kinase inhibition by the pyridinylfuranopyrimidine inhibitor PI-103 enhances tumor radiosensitivity. Cancer Res. 2008;68:5915–23.PubMedCrossRef Prevo R, Deutsch E, Sampson O, Diplexcito J, Cengel K, Harper J, et al. Class I PI3 kinase inhibition by the pyridinylfuranopyrimidine inhibitor PI-103 enhances tumor radiosensitivity. Cancer Res. 2008;68:5915–23.PubMedCrossRef
29.
Zurück zum Zitat Gachechiladze M, Skarda J, Soltermann A, Joerger M. RAD51 as a potential surrogate marker for DNA repair capacity in solid malignancies. Int J Cancer. 2017;141:1286–94.PubMedCrossRef Gachechiladze M, Skarda J, Soltermann A, Joerger M. RAD51 as a potential surrogate marker for DNA repair capacity in solid malignancies. Int J Cancer. 2017;141:1286–94.PubMedCrossRef
30.
Zurück zum Zitat Yoon SM, Ryoo B, Lee SJ, Kim JH, Shin JH, An JH, et al. Efficacy and safety of transarterial chemoembolization plus external beam radiotherapy vs sorafenib in hepatocellular carcinoma with macroscopic vascular invasion. JAMA Oncol. 2018;4:661–9.PubMedPubMedCentralCrossRef Yoon SM, Ryoo B, Lee SJ, Kim JH, Shin JH, An JH, et al. Efficacy and safety of transarterial chemoembolization plus external beam radiotherapy vs sorafenib in hepatocellular carcinoma with macroscopic vascular invasion. JAMA Oncol. 2018;4:661–9.PubMedPubMedCentralCrossRef
31.
Zurück zum Zitat Wang H, Mu X, He H, Zhang XD. Cancer radiosensitizers. Trends Pharmacol Sci. 2018;39:24–48.PubMedCrossRef Wang H, Mu X, He H, Zhang XD. Cancer radiosensitizers. Trends Pharmacol Sci. 2018;39:24–48.PubMedCrossRef
32.
Zurück zum Zitat Matsumoto S, Batra S, Saito K, Yasui H, Choudhuri R, Gadisetti C, et al. Antiangiogenic agent sunitinib transiently increases tumor oxygenation and suppresses cycling hypoxia. Cancer Res. 2011;71:6350–9.PubMedPubMedCentralCrossRef Matsumoto S, Batra S, Saito K, Yasui H, Choudhuri R, Gadisetti C, et al. Antiangiogenic agent sunitinib transiently increases tumor oxygenation and suppresses cycling hypoxia. Cancer Res. 2011;71:6350–9.PubMedPubMedCentralCrossRef
33.
Zurück zum Zitat Feng H, Cheng X, Kuang J, Chen L, Yuen S, Shi M, et al. Apatinib-induced protective autophagy and apoptosis through the AKT-mTOR pathway in anaplastic thyroid cancer. Cell Death Dis. 2018;9:1030.PubMedPubMedCentralCrossRef Feng H, Cheng X, Kuang J, Chen L, Yuen S, Shi M, et al. Apatinib-induced protective autophagy and apoptosis through the AKT-mTOR pathway in anaplastic thyroid cancer. Cell Death Dis. 2018;9:1030.PubMedPubMedCentralCrossRef
34.
Zurück zum Zitat Cheng X, Feng H, Wu H, Jin Z, Shen X, Kuang J, et al. Targeting autophagy enhances apatinib-induced apoptosis via endoplasmic reticulum stress for human colorectal cancer. Cancer Lett. 2018;431:105–14.PubMedCrossRef Cheng X, Feng H, Wu H, Jin Z, Shen X, Kuang J, et al. Targeting autophagy enhances apatinib-induced apoptosis via endoplasmic reticulum stress for human colorectal cancer. Cancer Lett. 2018;431:105–14.PubMedCrossRef
35.
36.
Zurück zum Zitat Bozulic L, Surucu B, Hynx D, Hemmings BA. PKBalpha/Akt1 acts downstream of DNA-PK in the DNA double-strand break response and promotes survival. Mol Cell. 2008;30:203–13.PubMedCrossRef Bozulic L, Surucu B, Hynx D, Hemmings BA. PKBalpha/Akt1 acts downstream of DNA-PK in the DNA double-strand break response and promotes survival. Mol Cell. 2008;30:203–13.PubMedCrossRef
37.
Zurück zum Zitat Toulany M, Lee KJ, Fattah KR, Lin YF, Fehrenbacher B, Schaller M, et al. Akt promotes post-irradiation survival of human tumor cells through initiation, progression, and termination of DNA-PKcs-dependent DNA double-strand break repair. Mol Cancer Res. 2012;10:945–57.PubMedCrossRef Toulany M, Lee KJ, Fattah KR, Lin YF, Fehrenbacher B, Schaller M, et al. Akt promotes post-irradiation survival of human tumor cells through initiation, progression, and termination of DNA-PKcs-dependent DNA double-strand break repair. Mol Cancer Res. 2012;10:945–57.PubMedCrossRef
38.
Zurück zum Zitat Leszczynska KB, Foskolou IP, Abraham AG, Anbalagan S, Tellier C, Haider S, et al. Hypoxia-induced p53 modulates both apoptosis and radiosensitivity via AKT. J Clin Invest. 2015;125:2385–98.PubMedPubMedCentralCrossRef Leszczynska KB, Foskolou IP, Abraham AG, Anbalagan S, Tellier C, Haider S, et al. Hypoxia-induced p53 modulates both apoptosis and radiosensitivity via AKT. J Clin Invest. 2015;125:2385–98.PubMedPubMedCentralCrossRef
39.
Zurück zum Zitat Ohri N, Dawson LA, Krishnan S, Seong J, Cheng JC, Sarin SK, et al. Radiotherapy for hepatocellular carcinoma: new indications and directions for future study. J Natl Cancer Inst. 2016;108:w133.CrossRef Ohri N, Dawson LA, Krishnan S, Seong J, Cheng JC, Sarin SK, et al. Radiotherapy for hepatocellular carcinoma: new indications and directions for future study. J Natl Cancer Inst. 2016;108:w133.CrossRef
40.
Zurück zum Zitat Clavreul A, Roger E, Pourbaghi-Masouleh M, Lemaire L, Tetaud C, Menei P. Development and characterization of sorafenib-loaded lipid nanocapsules for the treatment of glioblastoma. Drug Deliv. 2018;25:1756–65.PubMedPubMedCentralCrossRef Clavreul A, Roger E, Pourbaghi-Masouleh M, Lemaire L, Tetaud C, Menei P. Development and characterization of sorafenib-loaded lipid nanocapsules for the treatment of glioblastoma. Drug Deliv. 2018;25:1756–65.PubMedPubMedCentralCrossRef
41.
Zurück zum Zitat Jain RK. Normalization of tumor vasculature: an emerging concept in antiangiogenic therapy. Science. 2005;307:58–62.PubMedCrossRef Jain RK. Normalization of tumor vasculature: an emerging concept in antiangiogenic therapy. Science. 2005;307:58–62.PubMedCrossRef
43.
Zurück zum Zitat Liu Z, Wang Y, Huang Y, Kim BYS, Shan H, Wu D, et al. Tumor vasculatures: a new target for cancer immunotherapy. Trends Pharmacol Sci. 2019;40:613–23.PubMedCrossRefPubMedCentral Liu Z, Wang Y, Huang Y, Kim BYS, Shan H, Wu D, et al. Tumor vasculatures: a new target for cancer immunotherapy. Trends Pharmacol Sci. 2019;40:613–23.PubMedCrossRefPubMedCentral
44.
Zurück zum Zitat Faivre S, Raymond E, Boucher E, Douillard J, Lim HY, Kim JS, et al. Safety and efficacy of sunitinib in patients with advanced hepatocellular carcinoma: an open-label, multicentre, phase II study. Lancet Oncol. 2009;10:794–800.PubMedCrossRef Faivre S, Raymond E, Boucher E, Douillard J, Lim HY, Kim JS, et al. Safety and efficacy of sunitinib in patients with advanced hepatocellular carcinoma: an open-label, multicentre, phase II study. Lancet Oncol. 2009;10:794–800.PubMedCrossRef
45.
Zurück zum Zitat Cheng AL, Kang YK, Lin DY, Park JW, Kudo M, Qin S, et al. Sunitinib versus sorafenib in advanced hepatocellular cancer: results of a randomized phase III trial. J Clin Oncol. 2013;31:4067–75.PubMedCrossRef Cheng AL, Kang YK, Lin DY, Park JW, Kudo M, Qin S, et al. Sunitinib versus sorafenib in advanced hepatocellular cancer: results of a randomized phase III trial. J Clin Oncol. 2013;31:4067–75.PubMedCrossRef
46.
Zurück zum Zitat Yu W, Gu K, Yu Z, Yuan D, He M, Ma N, et al. Sorafenib potentiates irradiation effect in hepatocellular carcinoma in vitro and in vivo. Cancer Lett. 2013;329:109–17.PubMedCrossRef Yu W, Gu K, Yu Z, Yuan D, He M, Ma N, et al. Sorafenib potentiates irradiation effect in hepatocellular carcinoma in vitro and in vivo. Cancer Lett. 2013;329:109–17.PubMedCrossRef
47.
Zurück zum Zitat Hu C, Zhuang W, Qiao Y, Liu B, Liu L, Hui K, et al. Effects of combined inhibition of STAT3 and VEGFR2 pathways on the radiosensitivity of non-small-cell lung cancer cells. Onco Targets Ther. 2019;12:933–44.PubMedPubMedCentralCrossRef Hu C, Zhuang W, Qiao Y, Liu B, Liu L, Hui K, et al. Effects of combined inhibition of STAT3 and VEGFR2 pathways on the radiosensitivity of non-small-cell lung cancer cells. Onco Targets Ther. 2019;12:933–44.PubMedPubMedCentralCrossRef
Metadaten
Titel
Apatinib potentiates irradiation effect via suppressing PI3K/AKT signaling pathway in hepatocellular carcinoma
verfasst von
Junbin Liao
Huilin Jin
Shaoqiang Li
Lixia Xu
Zhenwei Peng
Guangyan Wei
Jianting Long
Yu Guo
Ming Kuang
Qi Zhou
Sui Peng
Publikationsdatum
01.12.2019
Verlag
BioMed Central
Erschienen in
Journal of Experimental & Clinical Cancer Research / Ausgabe 1/2019
Elektronische ISSN: 1756-9966
DOI
https://doi.org/10.1186/s13046-019-1419-1

Weitere Artikel der Ausgabe 1/2019

Journal of Experimental & Clinical Cancer Research 1/2019 Zur Ausgabe

Bei seelischem Stress sind Checkpoint-Hemmer weniger wirksam

03.06.2024 NSCLC Nachrichten

Wie stark Menschen mit fortgeschrittenem NSCLC von einer Therapie mit Immun-Checkpoint-Hemmern profitieren, hängt offenbar auch davon ab, wie sehr die Diagnose ihre psychische Verfassung erschüttert

Antikörper mobilisiert Neutrophile gegen Krebs

03.06.2024 Onkologische Immuntherapie Nachrichten

Ein bispezifischer Antikörper formiert gezielt eine Armee neutrophiler Granulozyten gegen Krebszellen. An den Antikörper gekoppeltes TNF-alpha soll die Zellen zudem tief in solide Tumoren hineinführen.

Erhebliches Risiko für Kehlkopfkrebs bei mäßiger Dysplasie

29.05.2024 Larynxkarzinom Nachrichten

Fast ein Viertel der Personen mit mäßig dysplastischen Stimmlippenläsionen entwickelt einen Kehlkopftumor. Solche Personen benötigen daher eine besonders enge ärztliche Überwachung.

15% bedauern gewählte Blasenkrebs-Therapie

29.05.2024 Urothelkarzinom Nachrichten

Ob Patienten und Patientinnen mit neu diagnostiziertem Blasenkrebs ein Jahr später Bedauern über die Therapieentscheidung empfinden, wird einer Studie aus England zufolge von der Radikalität und dem Erfolg des Eingriffs beeinflusst.

Update Onkologie

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