Skip to main content
Erschienen in: Radiation Oncology 1/2019

Open Access 01.12.2019 | Research

Stereotactic ablative radiotherapy versus conventionally fractionated radiotherapy in the treatment of hepatocellular carcinoma with portal vein invasion: a retrospective analysis

verfasst von: Jen-Fu Yang, Cheng-Hsiang Lo, Meei-Shyuan Lee, Chun-Shu Lin, Yang-Hong Dai, Po-Chien Shen, Hsing-Lung Chao, Wen-Yen Huang

Erschienen in: Radiation Oncology | Ausgabe 1/2019

Abstract

Background

This study aimed to compare the clinical outcomes of stereotactic ablative radiotherapy (SABR) and conventionally fractionated radiotherapy (CFRT) in hepatocellular carcinoma (HCC) patients with portal vein invasion (PVI).

Methods

HCC patients with PVI treated with radiotherapy from 2007 to 2016 were analysed. CFRT was administered at a median dose of 51.5 Gy (interquartile range, 45–54 Gy) with 1.8–3 Gy per fraction. SABR was administered at a median dose of 45 Gy (interquartile range, 40–48 Gy) with 6–12.5 Gy per fraction. Treatment efficacy, toxicity, and associated predictors were assessed.

Results

Among the 104 evaluable patients (45 in the SABR group and 59 in the CFRT group), the overall response rate (ORR, complete and partial response) was significantly higher in the SABR group than the CFRT group (62.2% vs. 33.8%, p = 0.003). The 1-year overall survival (OS) rate (34.9% vs. 15.3%, p = 0.012) and in-field progression-free survival (IFPS) rate (69.6% vs. 32.2%, p = 0.007) were also significantly higher in the SABR vs. CFRT group. All 3 rates remained higher in the SABR group after propensity score matching. Multivariable analysis identified SABR and a biologically effective dose ≥65 Gy as favourable predicators of OS. There was no difference between treatment groups in the incidence of radiation-induced liver disease or increase of Child-Pugh score ≥ 2 within 3 months of radiotherapy.

Conclusions

SABR was superior to CFRT in terms of ORR, OS, and IFPS. We suggest that SABR should be the preferred technique for HCC patients with PVI.
Hinweise

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Abkürzungen
3DCRT
Three dimensional conformal radiotherapy
AFP
Alpha-fetoprotein
BED
Biologically effective dose
CFRT
Conventionally fractionated radiotherapy
CP
Child-Pugh
CT
Computed tomography
CTV
Clinical target volume
ECOG
Eastern Cooperative Oncology Group
HCC
Hepatocellular carcinoma
IFPS
In-field progression-free survival
IQR
Inter-quartile range
MRI
Magnetic resonance imaging
ORR
Overall response rate
OS
Overall survival
PTV
Planning target volume
PVI
Portal vein invasion
PVT
Portal vein thrombosis
RILD
Radiation-induced liver disease
ROC
Receiver operating characteristic
RT
Radiotherapy
SABR
Stereotactic ablative radiotherapy
TACE
Transarterial chemoembolization

Background

Portal vein invasion (PVI) frequently develops in patients with advanced hepatocellular carcinoma (HCC), and has an estimated incidence rate of 34 to 80% [1, 2]. Without treatment, PVI prognosticates extremely poor prognosis with a median survival of only 2.7 months [3]. Sorafenib is currently regarded as the standard systemic therapy for HCC with PVI, but the survival gain is only 2–3 months [47].
For various locoregional modalities including surgical resection, transarterial chemoembolization (TACE), transarterial radioembolization, hepatic artery infusion chemotherapy attempted in patients with PVI, only carefully selected patients are amendable. Radiotherapy (RT) presents the only noninvasive alternative which is not dependent on vasculature to access the tumor, and is therefore not associated with a risk of hepatic ischemia. Over the past few decades, the development of the three-dimensional conformal technique has allowed for partial liver irradiation. Studies of conventionally fractionated radiotherapy (CFRT) in PVI have shown improved outcomes, with 1-year overall survival (OS) of 16.7–40.2% and overall response rates (ORR) of 23.5–45% [812].
Stereotactic ablative radiotherapy (SABR) is an emerging technique, and can achieve tumorcidal doses in limited fractions, with significant normal tissue sparing. Several studies have reported favorable results with SABR for treating different cancers [13, 14]. We hypothesized that SABR would provide more benefit than would CFRT in HCC patients with PVI.
The aim of this study was to present a single-institutional experience with a relatively large number of patients, and to compare the difference in clinical outcomes between the two RT techniques.

Methods

Patients

After obtaining waiver of consent for this retrospective research from the institutional review board of Tri-Service General Hospital (approval number: 1–107–05-016), we identified HCC patients with PVI undergoing RT from January 2007 to December 2016. Patients with Vp3/Vp4 invasion (invading the first-order branches and/or main trunk of the portal vein), an Eastern Cooperative Oncology Group (ECOG) performance status of 0 to 2 and a Child-Pugh (CP) class of A or B were eligible. Any prior interventions were allowed, except for previous RT. Those patients were considered unsuitable for standard locoregional treatment via multidisciplinary committee discussion. Given increasing evidence of the benefits of SABR for HCC patients in the past decades, our hospital practice naturally shifted from use of CFRT to use of SABR. Because SABR was not reimbursed until 2015 in Taiwan, the choice of RT technique was determined partly by the patients’ financial resources.
Diagnosis of HCC was made either by biopsy or by radiologic investigation based on characteristic imaging findings [15]. PVI was confirmed as a low-attenuation intraluminal mass that expanded the portal vein on contrast-enhanced helical computed tomography (CT) scans or on magnetic resonance imaging (MRI) scans. Pretreatment evaluation consisted of medical history, physical examination, complete blood counts, serum biochemistries, alpha-fetoprotein (AFP) level, chest film, and MRI and/or CT of the abdomen. Bone scan, positron emission tomography, or liver angiography was performed if clinically indicated.

CFRT technique

For better delineation of the upper gastrointestinal tract, most patients were asked to take oral contrast medium before simulation. A non-contrast CT simulation with a 3 mm slice thickness was performed. During the scanning procedure, all patients were supine, and were immobilized with a vacuum cushion, with arms raised overhead. Motion management with four-dimensional CT was conducted in some patients. Breath-holding was not mandatory. Either three dimensional conformal radiotherapy (3DCRT) or intensity-modulated radiotherapy was designated, depending on the physicians’ discretion, using the Nucletron Plato RTS v2.6.3 planning system, which was replaced by Philips Pinnacle v9.0 planning system in 2009. Instead of defining a gross target volume, the clinical target volume (CTV) was contoured directly. Contrast images registration was used to assist CTV delineation. The CTV was defined as a margin of 3–5 mm around the detectable PVI. Partial hepatic tumor was included in the CTV if the primary hepatic tumor was near to the detectable PVI or if the portal vein was invaded by the tumor directly. An example of target delineation is shown in Fig. 1. The planning target volume (PTV) was generated by expanding the CTV by 8 mm radially and by 10–15 mm craniocaudaully. Prescribed dose to the PTV was 45–54 Gy (1.8–3 Gy per fraction, five fractions per week), administered with a 15 MV photon beam. Most PTV was encompassed in the 90% isodose curve. The irradiation field contained 3–6 gantry angles. RT was delivered using an Elekta Precise or a Siemens Primus linear accelerator.

SABR technique

SABR was administered using the CyberKnife radiosurgery system (Accuray, Sunnyvale, CA), delivering 6 MV photons. At least one week before CT simulation, 4–6 fiducial markers were placed within or around the tumor under sonographic or CT guidance. An individually shaped vacuum pillow allowed patient immobilization in the supine position, with a vest for synchrony tracking and with abdominal compression devices to reduce respiratory motion. A contrast CT scan with a 1 mm slice thickness was obtained for treatment planning with or without MRI scan registration. The definition of CTV in SABR was the same as in the CFRT technique. The PTV was defined as the volume with a margin of 0–3 mm added to CTV for patients with fiducial implantation. If fiducial implantation failed or was unsuitable, the PTV margin was expanded to 8–20 mm craniocaudally, based on the liver motion. Margin modification was permitted for respecting normal tissue tolerance. Prior to August 2009, the MultiPlan CyberKnife treatment planning system version 1.7.0 was employed for treatment planning, and was updated to version 2.1.0 thereafter. The median total dose was 45 Gy, at 6–12.5 Gy per fraction, with 4 to 5 fractions administered on consecutive working days. The detailed dose-limiting organs and their constraints are described in our previous publication [16].

Evaluation and follow up

All patients were seen at least once per week during RT, 1–2 months for the first six months after RT, and every three months thereafter. Image evaluation with CT or MRI was obtained every 1–3 months. Modified Response Evaluation Criteria in Solid Tumors was used for evaluation of the PVI response [17].
Hepatic toxicity assessment consisted of Radiation-induced liver disease (RILD) and increase of CP score ≥ 2. Only patients with either adequate follow-up of three months or death and/or occurrence of toxicity within 3 months were included in the analysis. Physical examinations and blood tests conducted at every visit, were used for toxicity assessment. RILD was defined as either classic or non-classic, without intrahepatic tumor progression noted within three months after RT. Classic RILD manifested as the presence of nonmalignant ascites and the elevation of anicteric alkaline phosphatase level to at least twice the upper normal values. Non-classic RILD manifested as the elevation of transaminase levels to at least five times the upper limit of the normal or pre-treatment values.

Statistics

Doses were converted to biologically effective doses (BED) with an α/β ratio of 10 for analysis. Between-group comparisons were conducted by chi-square tests or by Student’s t-tests as appropriate. OS was measured from the first day of RT until the date of death from any cause or last follow-up. In-field progression-free survival (IFPS) was measured from the first day of RT until the date of tumor recurrence or progression in irradiated field or last follow-up and patients who have received local intervention (ex, surgery or reirradiation) were censored at the date of procedure. Kaplan Meier curves were constructed for OS and IFPS, and the difference was compared using the log-rank test. The Cox Proportional Hazards model was applied to identify potential predictors of survival. A receiver operating characteristic (ROC) curve was used to define optimal cut-off points for continuous variables based on the Youden index. Given the imbalance of potential confounders between groups, propensity score-matching was used. Propensity scores were estimated from a logistic regression model that included treatment period, sex, age, prior treatment, virus infection type, performance, tumor size, tumor number, extrahepatic metastasis, PVI location, AFP level, CP class, and sorafenib use. A nearest neighbor method with a caliper width of 0.2 was used to create matched cohorts. A 2-tailed p < 0.05 was designated as statistically significant for all tests except when variables with p < 0.10 in the univariable Cox model were entered into a multivariable Cox model. SPSS (SPSS Inc. Chicago, IL, USA) version 22 and R statistical software version 3.4.3 (R Foundation, https://​www.​r-project.​org)) were used for data analysis.

Results

Patient characteristics

Initially, 186 patients were identified by chart review. Forty-six patients were excluded owing to ineligibility, missing data or loss of follow-up after RT (Fig. 2). Finally, 140 patients were entered for data analysis. SABR was administered to 54 patients (median dose, 45 Gy; inter-quartile range [IQR], 40–48 Gy; 6–12.5 Gy per fraction), and CFRT was administered to 86 patients (median dose, 51.5; IQR, 45–54 Gy; 1.8–3 Gy per fraction). Detailed patient characteristics are listed in Table 1.
Table 1
Patient characteristics of the entire cohort
 
SABR (n = 54)
CFRT (n = 86)
  
Variable
No. of patients (%)
No. of patients (%)
p-value
SMD
Time of treatment
  
0.789
0.047
 Before December 31, 2011
27 (50.0)
45 (52.3)
  
 After December 31, 2011
27 (50.0)
41 (47.7)
  
Age, year
  
0.494
0.243
 Mean (SD)*, range
61.0 (12.9), 32–84
59.6 (11.2), 34–90
  
   ≤ 60
23 (42.6)
47 (54.7)
  
   > 60
31 (57.4)
39 (45.3)
  
Sex
  
0.766
0.052
 Male
42 (77.8)
65 (75.6)
  
 Female
12 (22.2)
21 (24.4)
  
Liver disease
  
0.281
0.086
 HBV
29 (53.7)
56 (65.1)
  
 HCV
17 (31.5)
16 (18.6)
  
 HBV and HCV
1 (1.9)
4 (4.7)
  
 Non-virus
7 (13.0)
10 (11.6)
  
ECOG
  
0.887
0.025
 0–1
46 (85.2)
74 (86.0)
  
 2
8 (14.8)
12 (14.0)
  
Extrahepatic metastasis
  
0.993
0.001
 Yes
5 (9.3)
8 (9.3)
  
 No
49 (90.7)
78 (90.7)
  
AFP, ng/ml
  
0.960
0.009
  ≤200
23 (42.6)
37 (43.0)
  
  > 200
31 (57.4)
49 (57.0)
  
Child-Pugh class
  
0.086
0.303
 A
35 (64.8)
43 (50.0)
  
 B
19 (35.2)
43 (50.0)
  
Prior treatment
  
0.047
0.350
 Yes
30 (55.6)
33 (38.4)
  
 No
24 (44.4)
53 (61.6)
  
No. of tumor
  
0.894
0.023
 Multiple
37 (68.5)
58 (67.4)
  
 Single
17 (31.5)
28 (32.6)
  
Tumor size, cm
  
0.088
0.298
  ≤8
28 (51.9)
32 (37.2)
  
  > 8
26 (48.1)
54 (62.8)
  
PVI location
  
0.128
0.267
 Vp4
23 (42.6)
48 (55.8)
  
 Vp3
31 (57.4)
38 (44.2)
  
Sorafenib
  
0.825
0.038
 Yes
23 (42.6)
35 (40.7)
  
 No
31 (57.4)
51 (59.3)
  
BED, Gy
  
< 0.001
1.075
  < 65
14 (25.9)
63 (73.3)
  
  ≥ 65
40 (74.1)
23 (26.7)
  
Abbreviations: SABR Stereotactic ablative radiotherapy, CFRT Conventionally fractionated radiotherapy, HBV Hepatitis B virus, HCV Hepatitis C virus, ECOG Eastern Cooperative Oncology Group, AFP Alpha fetoprotein, PVI Portal vein invasion, BED Biologically effective dose, SD Standard deviation, SMD Standardized mean difference. *t-test

PVI response

Follow-up images were available for 45 SABR patients and for 59 CFRT patients (83.3% vs. 68.6%, p = 0.052). Among evaluable patients, five complete response, 23 partial response, 15 stable disease, and two progressive disease cases were observed in the SABR group, and five complete response, 15 partial response, 27 stable disease, and 12 progressive disease cases were observed in the CFRT group. The ORR (complete and partial response) was significantly higher in the SABR group than it was in the CFRT group (62.2% vs. 33.9%, p = 0.004). Of all patients, 18 in the SABR group and 13 in the CFRT group were able to achieve either complete or partial recanalization of the invaded vein (33.3% vs. 15.1%, p = 0.012); subsequent TACE was conducted in 9 patients of the SABR group and in 11 patients of the CFRT group (16.7% vs. 12.8%, p = 0.524).

Survival

The median follow-up period was 6.2 months for all patients and 15.4 months for those alive. At the time of the analysis, nine patients in the SABR group and four patients in the CFRT group were alive. Before propensity score matching, the median survival was 10.9 months in the SABR group and 4.7 months in the CFRT group. The 1- and 2-year OS rates were 34.9% and 15.3% in the SABR group, and 15.7% and 8.0% in the CFRT group, respectively (p = 0.005, Fig. 3a). The 1- and 2-year IFPSs were also significantly higher in the SABR group compared to those in the CFRT group (69.6% vs. 39.8 and 32.2% vs. 24.2%, respectively; p = 0.007; Fig. 3b).
After propensity score-matching, 49 patients in each group were matched. (Table 2). The median survival was 10.7 months in the SABR group and 5.1 months in the CFRT group. The 1- and 2-year OS and IFPSs of the SABR group were significantly higher than those of the CFRT group (OS: 33.1% vs. 16.5% and 17.3% vs. 5.2%, p = 0.01, respectively; Fig. 4a; IFPS: 70.8% vs. 39.3% and 22.2% vs. 22.2%, p = 0.002, respectively; Fig. 4b).
Table 2
Patient characteristics of the propensity score-matched cohort
 
SABR (n = 49)
CFRT (n = 49)
  
Variable
No. of patients (%)
No. of patients (%)
p-value
SMD
Time of treatment
  
0.840
0.041
 Before December 31, 2011
24 (49.0)
23 (46.9)
  
 After December 31, 2011
25 (51.0)
26 (53.1)
  
Age, year
  
0.648
0.082
 Mean (SD)*, range
60.2 (13.2), 32–84
59.1 (10.9), 34–78
  
   ≤ 60
22 (44.9)
24 (49.0)
  
   > 60
27 (55.1)
25 (51.0)
  
Sex
  
0.812
0.048
 Male
38 (77.6)
37 (75.5)
  
 Female
11 (22.4)
12 (24.5)
  
Liver disease
  
0.259
0.116
 HBV
28 (57.1)
32 (65.3)
  
 HCV
14 (28.6)
7 (14.3)
  
 HBV and HCV
1 (2.0)
3 (6.1)
  
 Non-virus
6 (12.2)
7 (14.3)
  
ECOG
  
0.790
0.054
 0–1
41 (83.7)
40 (81.6)
  
 2
8 (16.3)
9 (18.4)
  
Extrahepatic metastasis
  
1.000
< 0.001
 Yes
5 (10.2)
5 (10.2)
  
 No
44 (89.8)
44 (89.8)
  
AFP, ng/ml
  
0.541
0.124
  ≤200
20 (40.8)
23 (46.9)
  
  > 200
29 (59.2)
26 (53.1)
  
Child-Pugh class
  
0.835
0.042
 A
31 (63.3)
30 (61.2)
  
 B
18 (36.7)
19 (38.8)
  
Prior treatment
    
 Yes
25 (51.0)
28 (57.1)
0.543
0.123
 No
24 (49.0)
21 (42.9)
  
No. of tumor
  
0.828
0.044
 Multiple
34 (69.4)
33 (67.3)
  
 Single
15 (30.6)
16 (32.7)
  
Tumor size, cm
  
1.000
< 0.001
  ≤8
24 (49.0)
24 (49.0)
  
  > 8
25 (51.0)
25 (51.0)
  
PVI location
  
1.000
< 0.001
 Vp4
21 (42.9)
21 (42.9)
  
 Vp3
28 (57.1)
28 (57.1)
  
Sorafenib
  
1.000
< 0.001
 Yes
21 (42.9)
21 (42.9)
  
 No
28 (57.1)
28 (57.1)
  
BED, Gy
  
< 0.001
1.124
  < 65
13 (26.5)
37 (75.5)
  
  ≥ 65
36 (73.5)
12 (24.5)
  
Abbreviations: SABR Stereotactic ablative radiotherapy, CFRT Conventionally fractionated radiotherapy, HBV Hepatitis B virus, HCV Hepatitis C virus, ECOG Eastern Cooperative Oncology Group, AFP Alpha fetoprotein, PVI Portal vein invasion, BED Biologically effective dose, SD Standard deviation, SMD Standardized mean difference. *t-test

Predictors for survival

Univariable analysis revealed that the presence of SABR, ECOG 0–1, CP class A, single tumor, tumor size ≤8 cm, Vp3, AFP ≤200 ng/ml, prior treatment, and BED ≥65 Gy were predictors of superior OS. No survival difference was noted between patients treated before December 31, 2011 versus after December 31, 2011. Given the presence of the high correlation between BED and the RT technique (p < 0.001), these variables were analyzed by two different Cox models to avoid collinearity. The presence of SABR and BED ≥65 Gy correlated significantly with superior OS in separate multivariable analysis models (Table 3). Furthermore, we identified that the SABR group with BED ≥65Gy showed a higher survival rate than the CFRT group with BED < 65Gy (p = 0.005) (Fig. 5).
Table 3
Univariable and multivariable analysis of the entire cohort
 
Univariable
Multivariable (model 1) †
Multivariable (model 2) †
Variable
HR (95% CI)
p
HR (95% CI)
p
HR (95% CI)
p
SABR vs. CFRT
0.615 (0.410–0.924)
0.019
0.623 (0.427–0.909)
0.014
  
BED < 65 vs. ≥ 65
1.482 (1.039–2.115)
0.030
  
1.682 (1.150–2.462)
0.007
Time of treatment Before December 31, 2011 vs. after December 31, 2011
1.008 (0.706–1.439)
0.965
    
Age ≤ 60 vs. > 60
0.882 (0.586–1.327)
0.547
    
Sex male vs. female
1.094 (0.687–1.741)
0.706
    
ECOG 0–1 vs. 2
0.554 (0.315–0.977)
0.041
0.523 (0.312–0.877)
0.014
0.572 (0.342–0.957)
0.033
Extrahepatic metastasis yes vs. no
1.731 (0.860–3.484)
0.124
    
AFP ≤200 vs. > 200
0.606 (0.424–0.866)
0.006
0.566 (0.378–0.846)
0.006
0.520 (0.344–0.785)
0.002
Child-Pugh class A vs. B
0.383 (0.249–0.588)
< 0.001
0.560 (0.381–0.823)
0.003
0.511 (0.345–0.756)
0.001
Prior Treatment yes vs no
0.690 (0.483–0.986)
0.042
0.955 (0.647–1.410)
0.816
0.903 (0.607–1.343)
0.615
Single vs. multiple
0.379 (0.233–0.617)
< 0.001
0.494 (0.323–0.756)
0.001
0.474 (0.308–0.729)
0.001
Tumor size ≤8 vs. > 8
0.452 (0.296–0.689)
< 0.001
0.538 (0.360–0.804)
0.002
0.550 (0.367–0.825)
0.004
Vp3 vs. Vp4
0.641 (0.426–0.964)
0.033
0.678 (0.467–0.985)
0.041
0.685 (0.471–0.995)
0.047
Sorafenib yes vs. no
0.980 (0.685–1.401)
0.911
    
Abbreviations: SABR Stereotactic ablative radiotherapy, BED Biologically effective dose, ECOG Eastern Cooperative Oncology Group, EM Extrahepatic metastasis, AFP Alpha feto protein, HR Hazard ratio, CI Confidence interval
†BED correlated highly with radiotherapy technique. Two Cox models were used to avoid collinearity

Toxicity

Acute toxicities observed in the groups are shown in Table 4. Fatigue was the most common adverse event in both groups. Grade 3 abdominal pain (n = 1) in the SABR group, and grade 3 diarrhea (n = 2) in the CFRT group were recorded. One patient in CFRT group presented with a treatment-related grade 5 duodenal ulcer.
Table 4
Acute toxicity observed due to SABR and CFRT in the entire cohort
 
SABR
CFRT
No. of patients (%)
No. of patients (%)
Grade 1
Grade 2
Grade 3
Grade 1
Grade 2
Grade 3
Grade 5
Nausea
5(9.3)
2(3.7)
 
9(10.5)
3(3.5)
  
Vomiting
3(5.6)
1(1.8)
 
2(2.3)
1(1.2)
  
Abdominal pain
9 (16.7)
2(3.7)
1(1.9)
6(7.0)
   
Diarrhea
1(1.9)
1(1.9)
 
6(7.0)
4(4.7)
2(2.3)
 
Fatigue
11 (20.3)
2(3.7)
 
17 (19.8)
1(1.2)
  
Anorexia
10 (18.5)
2(3.7)
 
10 (11.6)
1(1.2)
  
Duodenal ulcer
 
2(3.7)
  
1(1.2)
 
1(1.2)
Abbreviations: SABR Stereotactic ablative radiotherapy, CFRT Conventionally fractionated radiotherapy
Seven patients (13%) in the SABR group and 11 patients (12.8%) in the CFRT group experienced non-classic RILD, while two patients (3.7%) in the SABR group and six patients (7%) in the CFRT group experienced classic RILD. The incidences of RILD were not different between groups, even after pooling RILD types (16.7% vs. 19.8%, p = 0.646). There were no RILD–related deaths. After excluding 23 patients with missing follow-up CP scores, 10 patients (22.2%) in the SABR group and 19 patients (26.4%) in the CFRT group experienced an increase of CP score ≥ 2, which was not statistically different between groups (p = 0.612).

Discussion

Although CFRT has benefits in PVI, the long-term outcome is still poor. Thus, more effective and promising treatments like SABR need to be explored. Unlike previous CFRT series, most SABR series have included few or no patients with PVI; the efficacy of SABR in PVI is thus not clear. A few SABR series including only PVI patients have reported a 1-year OS of 43.2–50.3% and an ORR of 44.4–86.3% [1820]. Bujold et al. [21] have conducted the largest prospective trial of SABR for advanced HCC with 112 patients, of whom 55% presented with portal vein thrombosis (PVT). The overall 1-year OS was 44%, but the outcomes of the PVT subgroup were not reported.
Until now, direct head-to-head comparisons between different RT techniques in HCC with PVI have remained rare and controversial. In a cohort of patients with either PVT or inferior vena cava tumor thrombosis, Matsuo et al. [22] treated 43 patients with SABR (27 with CyberKnife and 16 with TrueBeam) and 54 patients with CFRT. The 1-year OS rate with SABR (using CyberKnife) was significantly higher than that with CFRT (56.7% vs. 29.3%, p = 0.02); similar trends were observed with local control and tumor response. In the meta-analysis performed by Rim et al. [23], SABR did not improve survival rates relative to CFRT in PVT patients. With only single-arm studies enrolled in their analysis, pooled estimates may not reveal the true head-to-head comparison as the heterogeneous designs and populations among studies. In the present study, the survival rates appear to be inferior to the published pooled results [23], in which the tumor size is much smaller than our cohort, with a median tumor size of 1.5–2.5 cm in the published pooled SABR cohort when compared with the median tumor size of 7.8 cm in our SABR cohort. However, when we analyzed only the outcome of tumors with ≤8 cm (median size: 5.1 cm) in our SABR series, the 1-year OS was 50% (data not shown), which is comparable to the 1-year OS of 48.5% in the pooled results. Tumor characteristics might partly explain the survival difference. Since some other factors affect survival as well, propensity score matching in the present study showed that SABR was superior to CFRT in terms of ORR, OS, and IFPS.
Some studies reported a higher prescribed dose or BED was correlated with better LC or survival [2426]. According to our multivariable model, a BED ≥65 Gy was found to be a predictor of survival prolongation. We further used a ROC curve to define optimal cut-off points for separate groups. Interestingly, higher BED was associated with better survival and ORR in the CFRT group, while no optimal cut-off point was found in the SABR group. In a CFRT series, Kim et al. [27] have reported that PVT patients receiving BED ≥58 Gy had a higher response rate than did those receiving BED < 58 Gy (54.6% vs. 20%, p = 0.034). Toya et al. [9] have also reported that BED (< 58 vs. ≥58 Gy) was a significant predictor of tumor response and survival. In one pooled analysis of SABR, Ohri et al. [28] reported that there was no dose response relationship in terms of local control when treating primary liver tumors, which is comparable to our findings. The narrow dose range and small sample size in our study may have led this result. We reasoned that higher objective response resulting from adequate BED contributes to survival benefit. In agreement, a previous hypothesis states that RT may reduce or stabilize PVI, leading to the restoration of vascular flow and slowing down of intrahepatic tumor dissemination, thus halting the deterioration of hepatic function.
Historically, RT has not been widely used for liver tumors because of a significant risk of RILD, though modern RT techniques have reduced this risk. A RILD incidence of 8–19% for CFRT [29, 30] and 0–5% for SABR [31, 32] has been reported. Moderately higher RILD incidence in our cohort may be explained by less favorable populations included, hepatitis B virus prevalence, and challenge to discern the causes of liver enzyme elevation. In agreement with our findings, CP score increase (another endpoint) was reported in 10 to 30% of primary liver cancer patients after SABR treatment [21, 33]. We chose these endpoints for robust data comparison. In our study, SABR and CFRT had comparable RILD incidences and CP score increases, which was possibly attributable to a relatively small cohort or the similar constraints (with 700 ml normal liver < 15 Gy) applied in both techniques. Even so, SABR leads to an improvement in therapeutic index, as it gives a greater level of clinical benefit for the same level of morbidity.
The high possibility of failure outside the radiation field implies the necessities for combining regional or systemic treatments with RT. Numerous combinations have been reported, though most were retrospective and poorly-evidenced. A recently-published randomized-controlled trial conducted by Yoon et al. [34] demonstrated that compared to sorafenib, TACE+3DCRT yielded higher progression-free survival, time to progression, ORR, and OS in HCC with macroscopic vascular invasion. We were unable to detect differences between TACE+SABR versus TACE+CFRT in subgroup analysis in our study. Given the potential superiority of SABR, replacement of CFRT with SABR is a consideration for future combination investigations.
The primary limitation of this study is the retrospective, single-institutional design. Even though propensity score matching minimizes the bias related to treatment assignment, unmeasured confounding may have existed. Further prospective studies are warranted. We note that issues regarding state-of-the-art technology may have affected treatment outcomes. However, we grouped patients into 2 treatment periods in the statistical analysis, which may eliminate this concern.

Conclusion

In summary, we demonstrated that compared to CFRT, SABR led to superior ORR, OS, and IFPS in propensity score-matched PVI patients. SABR delivers higher BED without increasing hepatic toxicities, and hence is a suitable RT modality for PVI patients. Further studies are required to validate our results.

Acknowledgements

We thank the Cancer Registry of the Tri-Service General Hospital for helping in data collection.
Waiver of consent for this retrospective research was obtained from the institutional review board of Tri-Service General Hospital (approval number: 1–107–05-016).
Not applicable.

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 Pirisi M, Avellini C, Fabris C, Scott C, Bardus P, Soardo G, et al. Portal vein thrombosis in hepatocellular carcinoma: age and sex distribution in an autopsy study. J Cancer Res Clin Oncol. 1998;124:397–400.CrossRef Pirisi M, Avellini C, Fabris C, Scott C, Bardus P, Soardo G, et al. Portal vein thrombosis in hepatocellular carcinoma: age and sex distribution in an autopsy study. J Cancer Res Clin Oncol. 1998;124:397–400.CrossRef
2.
Zurück zum Zitat Stuart KE, Anand AJ, Jenkins RL. Hepatocellular carcinoma in the United States. Prognostic feature, treatment outcome, and survival. Cancer. 1996;77:2217–22.CrossRef Stuart KE, Anand AJ, Jenkins RL. Hepatocellular carcinoma in the United States. Prognostic feature, treatment outcome, and survival. Cancer. 1996;77:2217–22.CrossRef
3.
Zurück zum Zitat Llovet JM, Bustamante J, Castells A, Vilana R, Ayuso Mdel C, Sala M, et al. Natural history of untreated nonsurgical hepatocellular carcinoma: rationale for the design and evaluation of therapeutic trials. Hepatology. 1999;29:62–7.CrossRef Llovet JM, Bustamante J, Castells A, Vilana R, Ayuso Mdel C, Sala M, et al. Natural history of untreated nonsurgical hepatocellular carcinoma: rationale for the design and evaluation of therapeutic trials. Hepatology. 1999;29:62–7.CrossRef
5.
Zurück zum Zitat European Association for Study of Liver; European Organisation for Research and Treatment of Cancer EASL-EORTC clinical practice guidelines: management of hepatocellular carcinoma Eur J Cancer 2012;48:599–641. European Association for Study of Liver; European Organisation for Research and Treatment of Cancer EASL-EORTC clinical practice guidelines: management of hepatocellular carcinoma Eur J Cancer 2012;48:599–641.
6.
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.CrossRef 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.CrossRef
7.
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.CrossRef 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.CrossRef
8.
Zurück zum Zitat Zeng ZC, Fan J, Tang ZY, Zhou J, Qin LX, Wang JH, et al. A comparison of treatment combinations with and without radiotherapy for hepatocellular carcinoma with portal vein and/or inferior vena cava tumor thrombus. Int J Radiat Oncol Biol Phys. 2005;61:432–43.CrossRef Zeng ZC, Fan J, Tang ZY, Zhou J, Qin LX, Wang JH, et al. A comparison of treatment combinations with and without radiotherapy for hepatocellular carcinoma with portal vein and/or inferior vena cava tumor thrombus. Int J Radiat Oncol Biol Phys. 2005;61:432–43.CrossRef
9.
Zurück zum Zitat Toya R, Murakami R, Baba Y, Nishimura R, Morishita S, Ikeda O, et al. Conformal radiation therapy for portal vein tumor thrombosis of hepatocellular carcinoma. Radiother Oncol. 2007;84:266–71.CrossRef Toya R, Murakami R, Baba Y, Nishimura R, Morishita S, Ikeda O, et al. Conformal radiation therapy for portal vein tumor thrombosis of hepatocellular carcinoma. Radiother Oncol. 2007;84:266–71.CrossRef
10.
Zurück zum Zitat Huang YF, Hsu HC, Wang CY, Wang CJ, Chen HC, Huang EY, et al. The treatment responses in cases of radiation therapy to portal vein thrombosis in advanced hepatocellular carcinoma. Int J Radiat Oncol Biol Phys. 2009;73:1155–63.CrossRef Huang YF, Hsu HC, Wang CY, Wang CJ, Chen HC, Huang EY, et al. The treatment responses in cases of radiation therapy to portal vein thrombosis in advanced hepatocellular carcinoma. Int J Radiat Oncol Biol Phys. 2009;73:1155–63.CrossRef
11.
Zurück zum Zitat Tanaka Y, Nakazawa T, Komori S, Hidaka H, Okuwaki Y, Takada J, et al. Radiotherapy for patients with unresectable advanced hepatocellular carcinoma with invasion to intrahepatic large vessels: efficacy and outcomes. J Gastroenterol Hepatol. 2014;29:352–7.CrossRef Tanaka Y, Nakazawa T, Komori S, Hidaka H, Okuwaki Y, Takada J, et al. Radiotherapy for patients with unresectable advanced hepatocellular carcinoma with invasion to intrahepatic large vessels: efficacy and outcomes. J Gastroenterol Hepatol. 2014;29:352–7.CrossRef
12.
Zurück zum Zitat Chen MY, Wang YC, Wu TH, Lee CF, Wu TJ, Chou HS, et al. Efficacy of external beam radiation-based treatment plus locoregional therapy for hepatocellular carcinoma associated with portal vein tumor thrombosis. Biomed Res Int. 2016;2016:6017406.PubMedPubMedCentral Chen MY, Wang YC, Wu TH, Lee CF, Wu TJ, Chou HS, et al. Efficacy of external beam radiation-based treatment plus locoregional therapy for hepatocellular carcinoma associated with portal vein tumor thrombosis. Biomed Res Int. 2016;2016:6017406.PubMedPubMedCentral
13.
Zurück zum Zitat Chang JY, Senan S, Paul MA, Mehran RJ, Louie AV, Balter P, et al. Stereotactic ablative radiotherapy versus lobectomy for operable stage I non-small-cell lung cancer: a pooled analysis of two randomised trials. Lancet Oncol. 2015;16:630–7.CrossRef Chang JY, Senan S, Paul MA, Mehran RJ, Louie AV, Balter P, et al. Stereotactic ablative radiotherapy versus lobectomy for operable stage I non-small-cell lung cancer: a pooled analysis of two randomised trials. Lancet Oncol. 2015;16:630–7.CrossRef
14.
Zurück zum Zitat Chuong MD, Springett GM, Freilich JM, Park CK, Weber JM, Mellon EA, et al. Stereotactic body radiation therapy for locally advanced and borderline resectable pancreatic cancer is effective and well tolerated. Int J Radiat Oncol Biol Phys. 2013;86:516–22.CrossRef Chuong MD, Springett GM, Freilich JM, Park CK, Weber JM, Mellon EA, et al. Stereotactic body radiation therapy for locally advanced and borderline resectable pancreatic cancer is effective and well tolerated. Int J Radiat Oncol Biol Phys. 2013;86:516–22.CrossRef
15.
Zurück zum Zitat Bruix J, Sherman M. American Association for the Study of Liver Diseases. Management of hepatocellular carcinoma: an update. Hepatology. 2011;53:1020–2.CrossRef Bruix J, Sherman M. American Association for the Study of Liver Diseases. Management of hepatocellular carcinoma: an update. Hepatology. 2011;53:1020–2.CrossRef
16.
Zurück zum Zitat Huang WY, Jen YM, Lee MS, Chang LP, Chen CM, Ko KH, et al. Stereotactic body radiation therapy in recurrent hepatocellular carcinoma. Int J Radiat Oncol Biol Phys. 2012;84:355–61.CrossRef Huang WY, Jen YM, Lee MS, Chang LP, Chen CM, Ko KH, et al. Stereotactic body radiation therapy in recurrent hepatocellular carcinoma. Int J Radiat Oncol Biol Phys. 2012;84:355–61.CrossRef
17.
Zurück zum Zitat Lencioni R, Llovet JM. Modified RECIST (mRECIST) assessment for hepatocellular carcinoma. Semin Liver Dis. 2010;30:52–60.CrossRef Lencioni R, Llovet JM. Modified RECIST (mRECIST) assessment for hepatocellular carcinoma. Semin Liver Dis. 2010;30:52–60.CrossRef
18.
Zurück zum Zitat Choi BO, Choi IB, Jang HS, Kang YN, Jang JS, Bae SH, et al. Stereotactic body radiation therapy with or without transarterial chemoembolization for patients with primary hepatocellular carcinoma: preliminary analysis. BMC Cancer. 2008;8:351.CrossRef Choi BO, Choi IB, Jang HS, Kang YN, Jang JS, Bae SH, et al. Stereotactic body radiation therapy with or without transarterial chemoembolization for patients with primary hepatocellular carcinoma: preliminary analysis. BMC Cancer. 2008;8:351.CrossRef
19.
Zurück zum Zitat Que JY, Lin LC, Lin KL, Lin CH, Lin YW, Yang CC. The efficacy of stereotactic body radiation therapy on huge hepatocellular carcinoma unsuitable for other local modalities. Radiat Oncol. 2014;9:120.CrossRef Que JY, Lin LC, Lin KL, Lin CH, Lin YW, Yang CC. The efficacy of stereotactic body radiation therapy on huge hepatocellular carcinoma unsuitable for other local modalities. Radiat Oncol. 2014;9:120.CrossRef
20.
Zurück zum Zitat Xi M, Zhang L, Zhao L, Li QQ, Guo SP, Feng ZZ, et al. Effectiveness of stereotactic body radiotherapy for hepatocellular carcinoma with portal vein and/or inferior vena cava tumor thrombosis. PLoS One. 2013;8:e63864.CrossRef Xi M, Zhang L, Zhao L, Li QQ, Guo SP, Feng ZZ, et al. Effectiveness of stereotactic body radiotherapy for hepatocellular carcinoma with portal vein and/or inferior vena cava tumor thrombosis. PLoS One. 2013;8:e63864.CrossRef
21.
Zurück zum Zitat Bujold A, Massey CA, Kim JJ, Brierley J, Cho C, Wong RK, et al. Sequential phase I and II trials of stereotactic body radiotherapy for locally advanced hepatocellular carcinoma. J Clin Oncol. 2013;31:1631–9.CrossRef Bujold A, Massey CA, Kim JJ, Brierley J, Cho C, Wong RK, et al. Sequential phase I and II trials of stereotactic body radiotherapy for locally advanced hepatocellular carcinoma. J Clin Oncol. 2013;31:1631–9.CrossRef
22.
Zurück zum Zitat Matsuo Y, Yoshida K, Nishimura H, Ejima Y, Miyawaki D, Uezono H, et al. Efficacy of stereotactic body radiotherapy for hepatocellular carcinoma with portal vein tumor thrombosis/inferior vena cava tumor thrombosis: evaluation by comparison with conventional three-dimensional conformal radiotherapy. J Radiat Res. 2016;57:512–23.CrossRef Matsuo Y, Yoshida K, Nishimura H, Ejima Y, Miyawaki D, Uezono H, et al. Efficacy of stereotactic body radiotherapy for hepatocellular carcinoma with portal vein tumor thrombosis/inferior vena cava tumor thrombosis: evaluation by comparison with conventional three-dimensional conformal radiotherapy. J Radiat Res. 2016;57:512–23.CrossRef
23.
Zurück zum Zitat Rim CH, Kim CY, Yang DS, Yoon WS. Comparison of radiation therapy modalities for hepatocellular carcinoma with portal vein thrombosis: a meta-analysis and systematic review. Radiother Oncol. 2018;129:112–22.CrossRef Rim CH, Kim CY, Yang DS, Yoon WS. Comparison of radiation therapy modalities for hepatocellular carcinoma with portal vein thrombosis: a meta-analysis and systematic review. Radiother Oncol. 2018;129:112–22.CrossRef
24.
Zurück zum Zitat Holliday EB, Tao R, Brownlee Z. Definitive radiation therapy for hepatocellular carcinoma with portal vein tumor thrombus. Clin Transl Radiat Oncol. 2017;4:39–45.CrossRef Holliday EB, Tao R, Brownlee Z. Definitive radiation therapy for hepatocellular carcinoma with portal vein tumor thrombus. Clin Transl Radiat Oncol. 2017;4:39–45.CrossRef
25.
Zurück zum Zitat Jang WI, Kim MS, Bae SH. High-dose stereotactic body radiotherapy correlates increased local control and overall survival in patients with inoperable hepatocellular carcinoma. Radiat Oncol. 2013;8:250.CrossRef Jang WI, Kim MS, Bae SH. High-dose stereotactic body radiotherapy correlates increased local control and overall survival in patients with inoperable hepatocellular carcinoma. Radiat Oncol. 2013;8:250.CrossRef
26.
Zurück zum Zitat Chadha AS, Gunther JR, Hsieh CE. Proton beam therapy outcomes for localized unresectable hepatocellular carcinoma. Radiother Oncol. 2019;133:54–61.CrossRef Chadha AS, Gunther JR, Hsieh CE. Proton beam therapy outcomes for localized unresectable hepatocellular carcinoma. Radiother Oncol. 2019;133:54–61.CrossRef
27.
Zurück zum Zitat Kim DY, Park W, Lim DH, Lee JH, Yoo BC, Paik SW, et al. Three-dimensional conformal radiotherapy for portal vein thrombosis of hepatocellular carcinoma. Cancer. 2005;103:2419–26.CrossRef Kim DY, Park W, Lim DH, Lee JH, Yoo BC, Paik SW, et al. Three-dimensional conformal radiotherapy for portal vein thrombosis of hepatocellular carcinoma. Cancer. 2005;103:2419–26.CrossRef
28.
Zurück zum Zitat Ohri N, Tomé WA, Méndez Romero A, Miften M, Ten Haken RK, Dawson LA, et al. Local control after stereotactic body radiation therapy for liver tumors. Int J Radiat Oncol Biol Phys. 2018. pii: S0360–3016(17)34525-X. Ohri N, Tomé WA, Méndez Romero A, Miften M, Ten Haken RK, Dawson LA, et al. Local control after stereotactic body radiation therapy for liver tumors. Int J Radiat Oncol Biol Phys. 2018. pii: S0360–3016(17)34525-X.
29.
Zurück zum Zitat Dawson LA, Normolle D, Balter JM, McGinn CJ, Lawrence TS, Ten Haken RK. Analysis of radiation-induced liver disease using the Lyman NTCP model. J Radiat Oncol Biol Phys. 2002;53:810–21.CrossRef Dawson LA, Normolle D, Balter JM, McGinn CJ, Lawrence TS, Ten Haken RK. Analysis of radiation-induced liver disease using the Lyman NTCP model. J Radiat Oncol Biol Phys. 2002;53:810–21.CrossRef
30.
Zurück zum Zitat Cheng JC, Wu JK, Lee PC, Liu HS, Jian JJ, Lin YM, et al. Biologic susceptibility of hepatocellular carcinoma patients treated with radiotherapy to radiation-induced liver disease. Int J Radiat Oncol Biol Phys. 2004;60:1502–9.CrossRef Cheng JC, Wu JK, Lee PC, Liu HS, Jian JJ, Lin YM, et al. Biologic susceptibility of hepatocellular carcinoma patients treated with radiotherapy to radiation-induced liver disease. Int J Radiat Oncol Biol Phys. 2004;60:1502–9.CrossRef
31.
Zurück zum Zitat Tse RV, Hawkins M, Lockwood G, Kim JJ, Cummings B, Knox J, et al. Phase I study of individualized stereotactic body radiotherapy for hepatocellular carcinoma and intrahepatic cholangiocarcinoma. J Clin Oncol. 2008;26:657–64.CrossRef Tse RV, Hawkins M, Lockwood G, Kim JJ, Cummings B, Knox J, et al. Phase I study of individualized stereotactic body radiotherapy for hepatocellular carcinoma and intrahepatic cholangiocarcinoma. J Clin Oncol. 2008;26:657–64.CrossRef
32.
Zurück zum Zitat Lee MT, Kim JJ, Dinniwell R, Brierley J, Lockwood G, Wong R, et al. Phase I study of individualized stereotactic body radiotherapy of liver metastases. J Clin Oncol. 2009;27:1585–91.CrossRef Lee MT, Kim JJ, Dinniwell R, Brierley J, Lockwood G, Wong R, et al. Phase I study of individualized stereotactic body radiotherapy of liver metastases. J Clin Oncol. 2009;27:1585–91.CrossRef
33.
Zurück zum Zitat Son SH, Choi BO, Ryu MR, Kang YN, Jang JS, Bae SH, et al. Stereotactic body radiotherapy for patients with unresectable primary hepatocellular carcinoma: dose-volumetric parameters predicting the hepatic complication. Int J Radiat Oncol Biol Phys. 2010;78:1073–80.CrossRef Son SH, Choi BO, Ryu MR, Kang YN, Jang JS, Bae SH, et al. Stereotactic body radiotherapy for patients with unresectable primary hepatocellular carcinoma: dose-volumetric parameters predicting the hepatic complication. Int J Radiat Oncol Biol Phys. 2010;78:1073–80.CrossRef
34.
Zurück zum Zitat Yoon SM, Ryoo BY, 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: a randomized clinical trial. JAMA Oncol. 2018;4:661–9.CrossRef Yoon SM, Ryoo BY, 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: a randomized clinical trial. JAMA Oncol. 2018;4:661–9.CrossRef
Metadaten
Titel
Stereotactic ablative radiotherapy versus conventionally fractionated radiotherapy in the treatment of hepatocellular carcinoma with portal vein invasion: a retrospective analysis
verfasst von
Jen-Fu Yang
Cheng-Hsiang Lo
Meei-Shyuan Lee
Chun-Shu Lin
Yang-Hong Dai
Po-Chien Shen
Hsing-Lung Chao
Wen-Yen Huang
Publikationsdatum
01.12.2019
Verlag
BioMed Central
Erschienen in
Radiation Oncology / Ausgabe 1/2019
Elektronische ISSN: 1748-717X
DOI
https://doi.org/10.1186/s13014-019-1382-1

Weitere Artikel der Ausgabe 1/2019

Radiation Oncology 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.