Skip to main content
Erschienen in: Discover Oncology 1/2024

Open Access 01.12.2024 | Clinical Trial

Transarterial chemoembolization with/without immune checkpoint inhibitors plus tyrosine kinase inhibitors for unresectable hepatocellular carcinoma: a single center, propensity score matching real-world study

verfasst von: Guosheng Yuan, Wenli Li, Mengya Zang, Rong Li, Qi Li, Xiaoyun Hu, Qi Zhang, Wei Huang, Jian Ruan, Huajin Pang, Jinzhang Chen

Erschienen in: Discover Oncology | Ausgabe 1/2024

Abstract

Objectives

To explore the efficacy and safety of Transarterial chemoembolization (TACE) in combination with immune checkpoint inhibitors (ICIs) and tyrosine kinase inhibitors (TKIs) in patients with unresectable hepatocellular carcinoma (uHCC).

Methods

456 patients with HCC receiving either TACE in combination with ICIs and TKIs (combination group, n = 139) or TACE monotherapy (monotherapy group, n = 317) were included from Apr 2016 to Dec 2021 in this retrospective study. We employed propensity score matching (PSM), performed 1:2 optimal pair matching, to balance potential bias.

Results

The mean follow-up time is 24.7 months (95% CI 22.6–26.8) for matched patients as of March 2022. After matching, the combination group achieved longer OS and PFS (median OS:21.9 vs. 16.3 months, P = 0.022; median PFS: 8.3 vs. 5.1 months, P < 0.0001) than TACE monotherapy group. The combination group had better objective response rate (ORR) and disease control rate (DCR) (ORR: 52.5% vs. 32.8%, P < 0.001; DCR: 82.7% vs. 59.6%, P < 0.001). Subgroup analysis showed that patients who received “TKIs + ICIs” after the first TACE procedure (after TACE group) achieved longer OS than those before the first TACE procedure (before TACE group) (26.8 vs. 19.2 months, P = 0.011). Adverse events were consistent with previous studies of TACE-related trials.

Conclusions

TACE plus TKIs and ICIs appeared to deliver longer PFS and OS in HCC patients than TACE monotherapy. “TKIs + ICIs” co-treatment within 3 months after the first TACE procedure might be a better medication strategy.
Hinweise

Supplementary Information

The online version contains supplementary material available at https://​doi.​org/​10.​1007/​s12672-024-00917-1.
Guosheng Yuan, Wenli Li and Mengya Zang contruibuted equally

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Abkürzungen
LC
Liver cancer
HCC
Hepatocellular carcinoma
TACE
Transarterial Chemoembolization
PSM
Propensity score matching
BCLC
Barcelona Clinic Liver Cancer
CNLC
China Liver Cancer Clinical
TME
Tumor microenvironment
ICIs
Immune checkpoint inhibitors
VEGF
Vascular endothelial growth factor
TKIs
Tyrosine kinase inhibitors
OS
Overall survival
PFS
Progression free survival
ORR
Objective response rate
DCR
Disease control rate
CR
Complete response
PR
Partial response
SD
Stable disease
PD
Progressive disease
AEs
Adverse events

1 Introduction

Liver cancer (LC) is one of the most prevalent causes of cancer-related death globally, especially hepatocellular carcinoma (HCC) associated with hepatitis B virus in Asia. [1] Although increasing rates of screening and surveillance for LC across most countries, more than 70% of patients are still diagnosed as intermediate or advanced-stage disease, leading to poor prognosis. [24]. TACE is widely used in intermediate HCC globally and in intermediate-advanced HCC domestically. [58] However, Transarterial chemoembolization (TACE) has not been universally successful in controlling tumor growth, recurrence and metastasis due to the high rate of incomplete embolization and embolization-induced changes in the tumor microenvironment (TME). [9, 10]
There are some rationales about TACE combining with immune checkpoint inhibitors (ICIs) and tyrosine kinase inhibitors (TKIs). Embolization-induced hypoxia after TACE can lead to upregulation of vascular endothelial growth factor (VEGF), which might contribute to revascularization, making it the theoretical basis of using anti-angiogenesis in the combination therapy. [11, 12] The efficacy of TACE in combination with anti-angiogenesis drugs in HCC has been proven in preclinical and clinical trials. [1315] In addition, TACE results in necrosis of the tumor tissue and releases tumor antigens which may promote tumor-specific immune responses. [16] It is reported that intra-tumoral exhausted effector cells, such as CD8 + /PD-1 + T cells and T regulatory cells (CD4 + /FOXP3 +) would become lower after TACE, which may transform an immuno-suppressive microenvironment into an immune supportive setting to enhance the response of ICIs. [17] Therefore, TACE combined with TKI and ICI is theoretically reasonable. To our knowledge, few data of prospective clinical trials are published about this regimen. Herein, more research is needed on the efficacy and safety of this triple combination therapy.
Therefore, we aimed to further validate the better efficacy with acceptable safety of TACE with ICI plus TKI than TACE monotherapy in this retrospective, propensity score matching (PSM) cohort of HCC patients in the real-world study.

2 Materials and methods

2.1 Patients

This retrospective study reviewed data of 854 unresectable HCC patients receiving TACE-based therapy from April 2016 to December 2021 (Fig. 1).
The inclusion criteria were as follows: (1) histologically, cytologically or clinically confirmed diagnosis of HCC according to the European Association for the Study of the Liver (EASL) criteria[5]; (2) at least 1 measurable lesion according to modified Response Evaluation Criteria in Solid Tumors (mRECIST, version 1.1); (3) with BCLC stage B or C; (4)received TACE or TACE + TKIs + ICIs as the first-line treatment, (5) received at least one cycle of TACE based therapy.
The exclusion criteria were as follows: (1) with Child–Pugh C liver function; (2) Eastern Cooperative Oncology Group (ECOG) score of ≥ 3 points; (3) liver transplantation, liver resection, or local–regional therapies before the TACE procedure; (4) currently had or had a history of malignant tumors in addition to HCC; (5) incomplete data;(6) not meeting the definition of combination therapy.
In the end, a total of 456 patients were selected for the study. Patients were assigned into 2 groups according to different treatment strategies: TACE monotherapy group and TACE in combination with TKIs and ICIs therapy group (combination group) (Fig. 1). Combination therapy was defined as treatment with TKIs and ICIs within 1 month before, or within 3 months after the first TACE procedure, while the interval between TKIs and ICIs must be within 1 week in the TACE + TKIs + ICIs group. To reduce the impact of potential confounders, we employed propensity score matching, performed 1:2 optimal pair matching to balance the covariates between the treatment groups (Table 1).
Table 1
Baseline characters of patients before and after PSM
Variables
Unmatched
Matched
TACE, N = 3171
TACE + TKIs + ICIs, N = 1391
p-value2
TACE, N = 2781
TACE + TKIs + ICIs, N = 1391
p-value2
Age(y)
  
0.708
  
0.942
Median (IQR)
59 (51, 66)
59 (50, 67)
 
58 (50, 67)
59 (50, 67)
 
Gender
  
0.144
  
0.315
Male
290 (91.48%)
121 (87.05%)
 
251 (90.29%)
121 (87.05%)
 
Female
27 (8.52%)
18 (12.95%)
 
27 (9.71%)
18 (12.95%)
 
BCLC
  
0.470
  
0.879
B
215 (67.82%)
99 (71.22%)
 
196 (70.50%)
99 (71.22%)
 
C
102 (32.18%)
40 (28.78%)
 
82 (29.50%)
40 (28.78%)
 
ECOG
  
0.408
  
0.776
0
259 (81.70%)
118 (84.89%)
 
233 (83.81%)
118 (84.89%)
 
1
58 (18.30%)
21 (15.11%)
 
45 (16.19%)
21 (15.11%)
 
Tumor Number
  
0.709
  
0.554
 ≥ 3 nodules
249 (78.55%)
107 (76.98%)
 
221 (79.50%)
107 (76.98%)
 
 < 3 nodules
68 (21.45%)
32 (23.02%)
 
57 (20.50%)
32 (23.02%)
 
Tumor Size(cm)
  
0.676
  
0.512
 < 7
109 (34.38%)
45 (32.37%)
 
99 (35.61%)
45 (32.37%)
 
 ≥ 7
208 (65.62%)
94 (67.63%)
 
179 (64.39%)
94 (67.63%)
 
Extrahepatic
metastasis
  
0.228
  
0.926
absent
249 (78.55%)
116 (83.45%)
 
231 (83.09%)
116 (83.45%)
 
present
68 (21.45%)
23 (16.55%)
 
47 (16.91%)
23 (16.55%)
 
PVTT
  
0.698
  
0.869
absent
241 (76.03%)
108 (77.70%)
 
214 (76.98%)
108 (77.70%)
 
present
76 (23.97%)
31 (22.30%)
 
64 (23.02%)
31 (22.30%)
 
AFP (ng/ml)
  
0.608
  
0.874
 < 400
242 (76.34%)
103 (74.10%)
 
208 (74.82%)
103 (74.10%)
 
 ≥ 400
75 (23.66%)
36 (25.90%)
 
70 (25.18%)
36 (25.90%)
 
Ascites
  
0.791
  
0.731
absent
252 (79.50%)
112 (80.58%)
 
220 (79.14%)
112 (80.58%)
 
present
65 (20.50%)
27 (19.42%)
 
58 (20.86%)
27 (19.42%)
 
Child–Pugh
  
0.903
  
0.922
A
270 (85.17%)
119 (85.61%)
 
237 (85.25%)
119 (85.61%)
 
B
47 (14.83%)
20 (14.39%)
 
41 (14.75%)
20 (14.39%)
 
WBC(*109)
  
0.746
  
0.878
Median (IQR)
5.21 (3.79, 6.67)
4.93 (3.95, 6.57)
 
5.23 (3.71, 6.64)
4.93 (3.95, 6.57)
 
NEU(*109)
  
0.684
  
0.880
Median (IQR)
2.91 (1.99, 4.05)
2.73 (2.00, 3.90)
 
2.88 (1.98, 3.99)
2.73 (2.00, 3.90)
 
PLT(*109)
  
0.451
  
0.515
Median (IQR)
141 (89, 208)
152 (101, 197)
 
141 (88, 214)
152 (101, 197)
 
ALT(U/L)
  
0.155
  
0.151
Median (IQR)
29 (21, 43)
32 (21, 48)
 
29 (21, 42)
32 (21, 48)
 
AST(U/L)
  
0.700
  
0.836
Median (IQR)
34 (24, 49)
32 (24, 47)
 
33 (24, 49)
32 (24, 47)
 
TBIL (µmol/L)
  
0.738
  
0.683
Median (IQR)
13 (9, 20)
13 (9, 18)
 
13 (9, 21)
13 (9, 18)
 
ALB(g/L)
  
0.889
  
0.996
Median (IQR)
37.5 (33.7, 41.0)
37.2 (34.0, 41.3)
 
37.6 (34.0, 40.9)
37.2 (34.0, 41.3)
 
PT(s)
  
0.478
  
0.282
Median (IQR)
11.90 (11.10, 12.80)
11.70 (11.00, 12.80)
 
11.90 (11.20, 12.90)
11.70 (11.00, 12.80)
 
TACE times
  
0.386
  
0.751
Median (IQR)
2.00 (1.00, 4.00)
2.00 (1.00, 3.00)
 
2.00 (1.00, 4.00)
2.00 (1.00, 3.00)
 
HBsAg
  
0.732
  
0.853
Positive
258 (81.39%)
115 (82.73%)
 
232 (83.45%)
115 (82.73%)
 
Negative
59 (18.61%)
24 (17.27%)
 
46 (16.55%)
24 (17.27%)
 
HBsAb
  
0.714
  
 > 0.999
Negative
279 (88.01%)
124 (89.21%)
 
248 (89.21%)
124 (89.21%)
 
Positive
38 (11.99%)
15 (10.79%)
 
30 (10.79%)
15 (10.79%)
 
HBeAg
  
0.558
  
0.670
Negative
282 (88.96%)
121 (87.05%)
 
246 (88.49%)
121 (87.05%)
 
Positive
35 (11.04%)
18 (12.95%)
 
32 (11.51%)
18 (12.95%)
 
HBeAb
  
0.565
  
0.781
Positive
139 (43.85%)
65 (46.76%)
 
126 (45.32%)
65 (46.76%)
 
Negative
178 (56.15%)
74 (53.24%)
 
152 (54.68%)
74 (53.24%)
 
HBcAg
  
0.277
  
0.613
Positive
215 (67.82%)
87 (62.59%)
 
181 (65.11%)
87 (62.59%)
 
negative
102 (32.18%)
52 (37.41%)
 
97 (34.89%)
52 (37.41%)
 
HBVDNA
  
0.390
  
0.573
Positive
121 (38.17%)
59 (42.45%)
 
110 (39.57%)
59 (42.45%)
 
Negative
196 (61.83%)
80 (57.55%)
 
168 (60.43%)
80 (57.55%)
 
HCVRNA
  
0.786
  
 > 0.999
Negative
305 (96.21%)
135 (97.12%)
 
269 (96.76%)
135 (97.12%)
 
Positive
12 (3.79%)
4 (2.88%)
 
9 (3.24%)
4 (2.88%)
 
TACE, transarterial chemoembolization; TKIs, tyrosine kinase inhibitors; ICIs, Immune checkpoint inhibitors; ECOG, Eastern Cooperative Oncology Group; AFP, α-fetoprotein; WBC, white blood cell; NEU, neutrophils; ALT, alanine aminotransferase; AST, aspartate aminotransferase; TBIL, total bilirubin; ALB, albumin; PT, prothrombin time; PLT, platelet count; HBsAg, hepatitis B surface antigen; Anti-HBs, hepatitis B s antibody; HBeAg, hepatitis B e antigen; Anti-HBe, hepatitis B e antibody; Anti-HBc, hepatitis B core antibody; HBVDNA, DNA of hepatitis B virus; HCVRNA, RNA of hepatitis C virus;
1n (%),2Wilcoxon rank sum test; Pearson's Chi-squared test; Fisher's exact test
This study was designed and performed according to the Declaration of Helsinki and was approved by the Medical Ethics Committee of Nanfang Hospital, Southern Medical University, and written, informed consent was obtained from each patient to retrospectively review and report on their medical records.

2.2 Treatment protocol

2.2.1 TACE procedures

The TACE protocol was performed as the previous study stated. Briefly, an emulsion mixed with lipiodol (5–20 mL) and adriamycin (20–50 mg) and subsequent 500–700 μm absorbable gelatin sponge particles were injected into the tumor feeding artery for chemoembolization.

2.2.2 The administration of TKIs

TKIs was given at a metronomic oral dosage (400 mg, twice daily, for sorafenib; 8 mg, daily, for lenvatinib; 80 mg, daily, for Regorafenib; 250 mg, daily, for Apatinib).

2.2.3 The administration of ICIs

Toripalimab was given at a fixed dose of 240 mg; Camrelizumab, Sintilimab and Tislelizumab was given at a fixed dose of 200 mg; Atezolizumab was given at a fixed dose of 1200 mg; Pembrolizumab was given at 2 mg/kg body weight; Nivolumab was given at 3 mg/kg body weight; All ICIs were given every 3 weeks intravenously.
The grading of adverse events related to TKIs, and ICIs were conducted according to the National Cancer Institute Common Terminology Criteria for Adverse Events (NCI-CTCAE 5.0). If adverse events ≥ 3 grade (defined as serious adverse events, SAEs) were observed, dosages of TKIs and ICIs were reduced, suspended, or discontinued.

2.3 Study endpoints

The primary endpoints of this study were OS (defined as the interval from treatment initiation (TACE or the first dose of TKIs/ICIs administration) to patient death or the last follow-up) and progression free survival (PFS) (defined as the time from treatment initiation to the first reported disease progression or death from any cause). The secondary endpoints were objective response rate (ORR) and disease control rate (DCR), according to the tumor response evaluated by two experienced radiologists using mRECIST. The ORR was defined as the percent of the total number of patients who had a complete response (CR) and partial response (PR), while DCR was defined as the percent of the total number of patients who had a CR, PR and stable disease (SD). Adverse events (AEs) were graded according to NCI-CTCAE 5.0. In addition, we had analyzed the predictors of affecting OS in the study. Whether the timing of TACE in combination with TKIs and ICIs affects OS was further explored in the TACE + TKIs + ICIs group.

2.4 Statistical analysis

To address the imbalance of potential confounders between two groups, we performed 1:2 optimal pair matching, propensity scores were calculated using logistic regression, taking into account the aforementioned demographic and clinical characteristic, including Age, Gender, BCLC, ECOG, Tumor Number, Tumor Size, Extrahepatic metastasis, PVTT, AFP, Ascites, Child–Pugh, WBC, NEU, ALT, AST, TBIL, ALB, PT, PLT, HBsAg, HBsAb, HBeAg, HBeAb, HBcAg, HBVDNA, and HCV, TACE time for both groups.
The patient characteristics were summarized using median with IQR for continuous variables and frequencies with proportions for categorical variables. Student’s t-test or Mann–Whitney U test was used to analyze continuous variables. Chi-squared test or Fisher exact test was applied to analyze categorical variables. The difference in PFS and OS between the two groups were compared with the use of a log-rank test. The survival curves were plotted by the method of Kaplan–Meier.
Cox proportion hazards model was used for multivariable analyses on the propensity-matched sample. Forest plots were used to display these data.
A two-tailed P-value of < 0.05 was considered statistically significant. All the above statistical analyses were performed using R (version 4.2.2; R Project for Statistical Computing, http://
www.​r-project.​org), MSTATA software and SPSS (version 25.0; IBM, SPSS).

3 Results

3.1 Patient baseline characteristics

A total of 456 patients were included in the final study: 317 (69.5%) patients received TACE monotherapy (TACE monotherapy group), and 139 (30.5%) patients received TACE in combination with TKIs and ICIs therapy (combination group), respectively. The mean follow-up time is 17.1 months (95% CI 16.3–17.9 months) and the mean number of TACE sessions were 2.7 (95% CI 2.5–2.9) for all patients as of March 2022. In the TACE monotherapy group, 102 (32.2%) patients were BCLC stage C, 259 (81.7%) had an ECOG performance score of 0, 249 (78.5%) had ≥ 3 tumor nodules, 76 (24.0%) were embolus existed; In the TACE + TKIs + ICIs group, 40 (28.8%) patients were BCLC stage C, 118 (84.9%) had an ECOG performance score of 0, 107 (77.0%) had ≥ 3 tumor nodules, 31 (22.3%) were embolus existed. There were no significant differences in baseline parameters between two groups both before and after matching (Table 1).

3.2 Efficacy

After matching, the median OS (mOS) and median PFS (mPFS) were both longer in TACE + TKIs + ICIs group than TACE alone with significant differences between the two group. The mOS were 21.9 months (95% CI 19.1–27.5 months) and 16.3 months (95% CI 14.5–21.0 months), respectively (P = 0.022). The mPFS were 8.3 months (95% CI 7.3–9.3 months) and 5.1 months (95% CI 4.5–6.2 months, respectively (P < 0.0001) (Fig. 2). More details about final status of patients are shown in Additional file 1: Table S2.
We used multivariableCox proportionalhazards model showed that combination therapy was the independent positive prognostic indicator for OS in the matched cohorts (hazard ratio [HR], 0.72,95% CI,0.54–0.95,P = 0.022 in univariable analysis; HR, 0.73,95% CI, 0.55–0.97,P = 0.032 in multivariable analysis), while Tumor Number is also another independent predictors of OS. (Table 2)Subgroup analysis of OS showed that the combination group had a trend persisted on better clinical benefits compared to the monotherapy group (Fig. 3). Additionally, we conducted Logistic regression analysis using progressive disease(PD) as a binary variable and found that the PD rate advantage of the TACE + TKIs + ICIs group is about 0.69 times that of the TACE group in the overall population (OR = 0.69, 95% CI 0.46–1.04, P = 0.079) (Additional file 1 Figure S1). At different subgroup levels, the OR values are always less than 1, implying a trend of lower rate of PD in the TACE + TKIs + ICIs than that of the TACE group.
Table 2
Predictors of overall survival after matching
 
Univariable
Multivariable
Characteristic
N
Event N
HR1
95% CI1
p-value
N
Event N
HR1
95% CI1
p-value
Gender
 Male
373
216
 
373
216
 
 Female
44
28
1.02
0.68, 1.51
0.935
44
28
1.25
0.81, 1.91
0.312
Age
417
244
1.00
0.99, 1.01
0.948
417
244
1.00
0.99, 1.01
0.745
BCLC
 B
288
154
 
288
154
 
 C
129
90
1.74
1.34, 2.27
 < 0.001
129
90
1.25
0.72, 2.17
0.428
ECOG
 0
342
199
 
342
199
 
 1
75
45
1.11
0.80, 1.53
0.546
75
45
1.09
0.78, 1.54
0.608
Tumor number
  ≥ 3 nodules
326
189
 
326
189
 
  < 3 nodules
91
55
1.43
1.06, 1.93
0.020
91
55
1.43
1.03, 1.98
0.031
Tumor Size
          
  < 7
139
83
 
139
83
 
  ≥ 7
278
161
1.13
0.86, 1.47
0.385
278
161
1.01
0.76, 1.34
0.962
Extrahepatic metastasis
 Absent
339
187
 
339
187
 
 Present
78
57
1.64
1.22, 2.21
0.001
78
57
1.49
1.00, 2.21
0.051
PVTT
          
 Absent
322
178
 
322
178
 
 Present
95
66
1.63
1.23, 2.17
 < 0.001
95
66
1.15
0.70, 1.91
0.579
AFP
  < 400
312
178
 
312
178
 
  ≥ 400
105
66
1.32
1.00, 1.76
0.053
105
66
1.17
0.87, 1.59
0.302
Child Pugh
 A
354
208
 
354
208
 
 B
63
36
0.93
0.65, 1.32
0.682
63
36
0.97
0.67, 1.41
0.881
Ascites
          
 Absent
329
189
 
329
189
 
 Present
88
55
1.14
0.84, 1.53
0.410
88
55
1.07
0.78, 1.48
0.656
HBVDNA
 Positive
168
103
 
168
103
 
 Negative
249
141
0.76
0.59, 0.99
0.038
249
141
0.80
0.60, 1.05
0.112
HCVRNA
 Negative
401
235
 
401
235
 
 Positive
16
9
1.24
0.64, 2.42
0.523
16
9
1.22
0.61, 2.47
0.571
Group
 TACE
278
171
 
278
171
 
 TACE + TKIs + ICIs
139
73
0.72
0.54, 0.95
0.022
139
73
0.73
0.55, 0.97
0.032
1HR = Hazard Ratio, CI = Confidence Interval
In the TACE monotherapy group, 6 (1.9%) patients had complete response (CR), 98 (30.9%) patients achieved PR, and 85 (26.8%) patients had SD, resulting in an ORR of 32.8% and DCR of 59.6%. In the TACE + TKIs + ICIs group, 9 (6.5) and 64 (46.0%) participants achieved CR and PR, respectively, with 42 (30.2%) patients experiencing SD and 24 (17.3%) experiencing PD. The ORR was 52.5%, and the DCR was 82.7% for this treatment group (P = 0.001 for ORR comparison; P < 0.001 for DCR comparison) (Table 3).
Table 3
Tumor responses
Tumor response, n (%)
TACE (n = 317)
TACE + TKIs + ICIs (n = 139)
Complete response (CR)
6 (1.9)
9 (6.5)
Partial response (PR)
98 (30.9)
64 (46.0)
Stable disease (SD)
85 (26.8)
42 (30.2)
Progressive disease (PD)
128 (40.4)
24 (17.3)
ORR (CR + PR)*
104 (32.8)
73 (52.5)
DCR (CR + PR + SD)#
189 (59.6)
115 (82.7)
DCR, disease control rate; ORR, objective response rate
*Pearson χ2 = 15.807, P < 0.001 (ORR comparison between the 2 groups)
#Pearson χ2 = 23.228, P < 0.001 (DCR comparison between the 2 groups)
multivariable.

3.3 Safety

Totally 248 patients (78.2%) in the TACE monotherapy group and 112 (80.6%) in the TACE + TKIs + ICIs group, experienced at least one AEs of any grade. The most common treatment-related AEs (TRAEs) were pain (68.8%), fever (51.4%), and thrombocytopenia (49.8%) in the TACE monotherapy group; Fatigue (62.6%), Aspartate aminotransferase increased (61.9%), and proteinuria (59.7%) in the TACE + TKIs + ICIs group.
The TRAEs ≥ 3 grade led to dose reduction in 36 (11.4%) patients and treatment termination in 2 (0.6%) patients in the TACE monotherapy group, and 30 (21.6%) patients and 8 (5.8%) patients in the TACE + TKIs + ICIs group. AEs in all patients were effectively controlled by symptomatic treatment or reduction in drug dose or discontinuation, and there were no treatment-related deaths (Table 4).
Table 4
Treatment-related adverse events
Effects
TACE (n = 317)
TACE + TKIs + ICIs (n = 139)
P value
Alanine aminotransferase increased
134 (42.3)
79 (56.8)
0.004
Alimentary tract hemorrhage
16 (5.0)
13 (9.4)
0.083
Aspartate aminotransferase increased
143 (45.1)
86 (61.9)
0.001
Diarrhea
92 (29.0)
51 (36.7)
0.104
Fatigue
114 (36.0)
87 (62.6)
 < 0.001
Fever
163 (51.4)
77 (16.5)
0.434
Gingivitis
62 (19.6)
23 (16.5)
0.447
Hand-foot syndrome
41 (12.9)
72 (51.8)
 < 0.001
Hematuria
34 (10.7)
19 (13.7)
0.367
Hyperbilirubinemia
22 (6.9)
42 (30.2)
 < 0.001
Hypertension
17 (5.4)
53 (38.1)
 < 0.001
HyperthyroidismΔ
8 (2.5)
5 (3.6)
0.547
HypothyroidismΔ
4 (1.3)
6 (4.3)
0.074
Nausea
117 (36.9)
67 (48.2)
0.024
Pain
218 (68.8)
71 (51.1)
 < 0.001
Proteinuria
14 (4.4)
83 (59.7)
 < 0.001
Rash
76 (24.0)
53 (38.1)
0.002
Thrombocytopenia
158 (49.8)
82 (59.0)
0.072
Weight loss
37 (11.7)
71 (51.1)
 < 0.001
Grade ≥ 3
36 (11.4)
30 (21.6)
0.007
Dose reduction
2 (0.6)
8 (5.8)
0.002
Δ: Fisher's exact test, others used χ2 tests

3.4 Subgroup analysis of “before TACE group” and “after TACE group”

To determine the timing of TACE in combination with TKIs and ICIs, the mOS was compared between the “before TACE group” (the “TKIs + ICIs” co-treatment was given within 1 month before the first TACE procedure) and the “after TACE group” (the “TKIs + ICIs” co-treatment was given within 3 months after the first TACE procedure). According to Figure S1 in Additional file 1, the mOS was 19.2 months (95% CI 15.3–25.0 months) in the “before TACE group” and 26.8 months (95% CI 20.2–45.1 months) in the “after TACE group” (P = 0.011).(Fig. 4).

4 Discussion

Our study showed that TACE in combination with TKIs and ICIs achieved the higher mOS (21.9 months, 95% CI 17.2–26.6 months) and mPFS (8.3 months, 95% CI 6.0–7.4 months) when compared to TACE alone. Subgroup analyses showed that the mOS was 19.2 months (95% CI 14.3–24.1 months) in the “before TACE group” and 26.8 months (95% CI 15.7–37.9 months) in the “after TACE group” (P = 0.011). No worse AEs were observed in the present study compared with previously reported clinical trials. [1820] Most AEs were easily managed and controlled with mild-to-moderate severity, consistent with the results of other previous studies. This combination regimen promises to be a potential first-line option for patients with advanced HCC.
Currently, immunotherapy has approved for the treatment in HCC. “A + T” has been recommended as the first-line treatment in andvanced HCC after IMbrave150 trial published. Compared to updated data of IMbrave150 and Oriental-32, our study seemed to delivery more promising mOS (21.9 vs 19.2 compared IMbrave150, 21.9 vs 10.4 compared Orient-32) and mPFS (8.3 vs 6.9 compared IMbrave150, 8.3 vs 6.9 compared Orient-32). [21, 22] It probably means that combining TACE with TKIs + ICIs improves mOS and mPFS of patients with intermediate-advanced HCC. This has also been validated in real-world studies. Median OS and PFS of combination group were 19.2 months [95%CI (16.1–27.3)] and 9.5 months [95%CI (8.4–11.0)] in CHANCE001 trial, 24.1 months [95%CI 20.0-NR] and 13.5 months [95%CI 9.0–18.0] in CHANCE2211 trial, respectively. [23, 24] Our study demonstrated considerable efficacy, especially in “after TACE” group. However, the ORR of immunotherapy in HCC patients is 15–30%. Its efficacy is affected by a variety of factors, such as liver function status, BCLC staging, baseline tumor burden, etiology, etc. [25, 26] How to improve the efficacy of immunotherapy is still a problem for clinicians. The OS benefit with atezolizumab plus bevacizumab vs. sorafenib was generally consistent across the patient subgroups in IMbrave150 trial, except in patients who had non-viral etiology of HCC (HR for death 1.05; 95% CI 0.68–1.63). [25, 27] Immune therapy did not improve survival in patients with non-viral HCC reported in a meta-analysis, which showed that non-viral HCC, might be less responsive to immunotherapy. That was visibly observed in HCC caused by Non-Alcoholic SteatoHeaptitis(NASH-HCC) probably owing to NASH-related aberrant T cell activation causing tissue damage and impaired immune surveillance. [25, 28]While the synergistic antitumor effects underlying TACE plus ICIs and TKIs was verified and could possibly be explained as: (1) TACE causes necrosis and disintegration of the tumor cells, which results in releasing a large number of tumor antigens, increasing the expression of programmed cell death protein-1 (PD-1) and programmed cell death protein ligand-1 (PD-L1), and improving tumor recognition [12, 17]; (2) TACE could alter the TME of HCC by producing local inflammation, promoting lymphocyte infiltration, and activating the immune system, which may improve the overall anticancer effect of ICIs [16, 29]; (3) TKIs could antagonize the effect of TACE-induced angiogenesis by blocking the VEGF signaling pathway and enhance the efficacy of ICIs by reversing VEGF-mediated immunosuppression [3032]. These proposed mechanisms suggest that the combination of TACE, TKIs and ICIs might produce synergistic anti-HCC effects and therefore improve the OS and PFS of the patients.
In the subgroup analysis, “After TACE group” delivered more promising result of OS compared “Before TACE group”. One possible explanation for prolong mOS is that the efficacy of TACE, tumor antigens releasing and TME altering, are time-consuming [33], possibly due to the worsened hypoxia and immunosuppressive TME of residual tumor tissue following TACE [9, 32]. At this consuming moment, TKIs are needed to alleviate hypoxia and immunosuppressive TME through vascular normalization so that the inflammatory environment suitable for T-cell responses could be maintained, which improves the therapeutic effectiveness of ICIs [31]. In addition, previous studies have found a significant infiltration of CD8 + T cells after TACE, indicating an immunological enhancement in the body [12, 34, 35]. LAUCH trial displayed better benefits of TACE plus lenvatinib than TACE monotherapy, which means TKIs administration after TACE procedure may improve the efficacy of TACE. The rationale is clear to conduct ICIs treatment on the basis of inflammatory response and enhanced immunity after TACE.
Compared with monotherapy group, worse liver function, infusion-related reaction, proteinuria, pain and Hand-foot syndrome were observed in combination group. Fortunately, all adverse events were controlled and manageable with dosage reduction. Lower incidence rate of grade 3 or higher AEs occurred than those reported in IMbrave150, ORIENT-32 and CARES-310 trials (21.6% vs 63%, 21.6% vs 56% and 21.6% vs 71%). [21, 36, 37]Grade 3 or higher AEs in our study is similar to other real word studies (16.7% in CHANCE2211 and 15.8% in CHANCE2201). [23, 24] Safety is one of challenges in combination therapy. Collectively, TACE + TKIs + ICIs therapy had acceptable safety and improved the OS and PFS of patients.
There were several limitations in this study. Firstly, this was a retrospectively designed study, which may lead to potential selection bias. Besides, the sample size of the study is limited. The conclusions drawn from this study should be verified in better-designed prospective studies. Thirdly, a group of patients taking TKIs before TACE but accepting ICIs therapy after TACE was lacking, since several studies found that pre-treatment with TKIs before TACE can normalize tumor vessels and suppress VEGF, which may lead to a homogeneous distribution of lipiodol mixed anticancer drugs in the tumors [15, 36, 37]. This might be an issue to be further explored. Additionally, another limitation of our study was a dropout rate of approximately 10%, but without different significance between the two groups. We had tried our best to make that within a statistically reasonable range, but we need research with lower or even without dropout to improve the reliability of the results in the future. Finally, although we used PSM to minimize bias as much as possible, the diversity of TKIs and ICIs can still lead to biased results. It makes sense that similar results in TACE plus mono-TKI and mono-ICI but more rigorously prospective studies needed to confirm this.
In conclusion, our retrospective data indicated that TACE + TKIs + ICIs seemed to have considerable efficacy in patients with unresectable HCC compared to TACE monotherapy. Specially, “TKIs + ICIs” co-treatment within 3 months after the first TACE procedure might be a better medication strategy to achieve better clinical outcomes.

Acknowledgements

This study was supported by grants from the National Natural Science Foundation of China (82102879), the Natural Science Foundation of Guangdong Province (2022A1515010526, 2021A1515012518), and the Postdoctoral Research Foundation of China (No. 2021M691468). The funding agencies had no role in the study design, data collection and analysis, decision to publish, or preparation of the manuscript.

Declarations

The authors are accountable for all aspects of the work in ensuring that questions related to the accuracy or integrity of any part of the work are appropriately investigated and resolved. This study was designed and performed in accordance with the Declaration of Helsinki and was approved by the Medical Ethics Committee of Nanfang hospital, Southern Medical University (NFEC-201903-Y1-01), and written, informed consent was obtained from each patient to retrospectively review and report on their medical records.

Competing interests

All authors have completed the ICMJE uniform disclosure form. The authors have no conflicts of interest to declare.
Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://​creativecommons.​org/​licenses/​by/​4.​0/​.

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Unsere Produktempfehlungen

e.Med Interdisziplinär

Kombi-Abonnement

Für Ihren Erfolg in Klinik und Praxis - Die beste Hilfe in Ihrem Arbeitsalltag

Mit e.Med Interdisziplinär erhalten Sie Zugang zu allen CME-Fortbildungen und Fachzeitschriften auf SpringerMedizin.de.

e.Med Innere Medizin

Kombi-Abonnement

Mit e.Med Innere Medizin erhalten Sie Zugang zu CME-Fortbildungen des Fachgebietes Innere Medizin, den Premium-Inhalten der internistischen Fachzeitschriften, inklusive einer gedruckten internistischen Zeitschrift Ihrer Wahl.

Literatur
1.
Zurück zum Zitat Sung H, et al. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J Clin. 2021;71:209–49.CrossRefPubMed Sung H, et al. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J Clin. 2021;71:209–49.CrossRefPubMed
4.
Zurück zum Zitat Yang JD, et al. A global view of hepatocellular carcinoma: trends, risk, prevention and management. Nat Rev Gastroenterol Hepatol. 2019;16:589–604.CrossRefPubMedPubMedCentral Yang JD, et al. A global view of hepatocellular carcinoma: trends, risk, prevention and management. Nat Rev Gastroenterol Hepatol. 2019;16:589–604.CrossRefPubMedPubMedCentral
5.
Zurück zum Zitat European Association for the Study of the Liver. Electronic address: easloffice@easloffice.eu & European Association for the Study of the Liver. EASL Clinical Practice Guidelines for the management of patients with decompensated cirrhosis. J Hepatol. 2018;69:406–60.CrossRef European Association for the Study of the Liver. Electronic address: easloffice@easloffice.eu & European Association for the Study of the Liver. EASL Clinical Practice Guidelines for the management of patients with decompensated cirrhosis. J Hepatol. 2018;69:406–60.CrossRef
6.
Zurück zum Zitat Xie D-Y, Ren Z-G, Zhou J, Fan J, Gao Q. 2019 Chinese clinical guidelines for the management of hepatocellular carcinoma: updates and insights. Hepatobiliary Surg Nutr. 2020;9:452–63.CrossRefPubMedPubMedCentral Xie D-Y, Ren Z-G, Zhou J, Fan J, Gao Q. 2019 Chinese clinical guidelines for the management of hepatocellular carcinoma: updates and insights. Hepatobiliary Surg Nutr. 2020;9:452–63.CrossRefPubMedPubMedCentral
7.
Zurück zum Zitat Rogal SS, Hansen L, Patel A, et al. AASLD practice guidance: palliative care and symptom-based managementindecompensatedcirrhosis. Hepatology. 2022;76(3):819–53.CrossRefPubMed Rogal SS, Hansen L, Patel A, et al. AASLD practice guidance: palliative care and symptom-based managementindecompensatedcirrhosis. Hepatology. 2022;76(3):819–53.CrossRefPubMed
8.
Zurück zum Zitat Reig M, et al. BCLC strategy for prognosis prediction and treatment recommendation: the 2022 update. J Hepatol. 2022;76:681–93.CrossRefPubMed Reig M, et al. BCLC strategy for prognosis prediction and treatment recommendation: the 2022 update. J Hepatol. 2022;76:681–93.CrossRefPubMed
9.
Zurück zum Zitat Wei X, et al. MiR-125b loss activated HIF1α/pAKT loop, leading to transarterial chemoembolization resistance in hepatocellular carcinoma. Hepatology. 2021;73:1381–98.CrossRefPubMed Wei X, et al. MiR-125b loss activated HIF1α/pAKT loop, leading to transarterial chemoembolization resistance in hepatocellular carcinoma. Hepatology. 2021;73:1381–98.CrossRefPubMed
10.
Zurück zum Zitat Raoul J-L, Gilabert M, Piana G. How to define transarterial chemoembolization failure or refractoriness: a european perspective. Liver Cancer. 2014;3:119–24.CrossRefPubMedPubMedCentral Raoul J-L, Gilabert M, Piana G. How to define transarterial chemoembolization failure or refractoriness: a european perspective. Liver Cancer. 2014;3:119–24.CrossRefPubMedPubMedCentral
11.
Zurück zum Zitat Chen M, et al. HIF-2α-targeted interventional chemoembolization multifunctional microspheres for effective elimination of hepatocellular carcinoma. Biomaterials. 2022;284: 121512.CrossRefPubMed Chen M, et al. HIF-2α-targeted interventional chemoembolization multifunctional microspheres for effective elimination of hepatocellular carcinoma. Biomaterials. 2022;284: 121512.CrossRefPubMed
12.
Zurück zum Zitat Llovet JM, et al. Locoregional therapies in the era of molecular and immune treatments for hepatocellular carcinoma. Nat Rev Gastroenterol Hepatol. 2021;18:293–313.CrossRefPubMed Llovet JM, et al. Locoregional therapies in the era of molecular and immune treatments for hepatocellular carcinoma. Nat Rev Gastroenterol Hepatol. 2021;18:293–313.CrossRefPubMed
13.
Zurück zum Zitat Kudo M, et al. Lenvatinib as an initial treatment in patients with intermediate-stage hepatocellular carcinoma beyond up-to-seven criteria and child-pugh a liver function: a proof-of-concept study. Cancers. 2019;11:1084.CrossRefPubMedPubMedCentral Kudo M, et al. Lenvatinib as an initial treatment in patients with intermediate-stage hepatocellular carcinoma beyond up-to-seven criteria and child-pugh a liver function: a proof-of-concept study. Cancers. 2019;11:1084.CrossRefPubMedPubMedCentral
14.
Zurück zum Zitat Geschwind J-F, et al. TACE treatment in patients with sorafenib-treated unresectable hepatocellular carcinoma in clinical practice: Final analysis of GIDEON. Radiology. 2016;279:630–40.CrossRefPubMed Geschwind J-F, et al. TACE treatment in patients with sorafenib-treated unresectable hepatocellular carcinoma in clinical practice: Final analysis of GIDEON. Radiology. 2016;279:630–40.CrossRefPubMed
15.
Zurück zum Zitat Kudo M, et al. Final results of TACTICS: a randomized, prospective trial comparing transarterial chemoembolization plus sorafenib to transarterial chemoembolization alone in patients with unresectable hepatocellular carcinoma. Liver Cancer. 2022;11:354–67.CrossRefPubMedPubMedCentral Kudo M, et al. Final results of TACTICS: a randomized, prospective trial comparing transarterial chemoembolization plus sorafenib to transarterial chemoembolization alone in patients with unresectable hepatocellular carcinoma. Liver Cancer. 2022;11:354–67.CrossRefPubMedPubMedCentral
16.
Zurück zum Zitat Chang X, Lu X, Guo J, Teng G-J. Interventional therapy combined with immune checkpoint inhibitors: Emerging opportunities for cancer treatment in the era of immunotherapy. Cancer Treat Rev. 2019;74:49–60.CrossRefPubMed Chang X, Lu X, Guo J, Teng G-J. Interventional therapy combined with immune checkpoint inhibitors: Emerging opportunities for cancer treatment in the era of immunotherapy. Cancer Treat Rev. 2019;74:49–60.CrossRefPubMed
17.
Zurück zum Zitat Montasser A, et al. Transarterial chemoembolisation enhances programmed death-1 and programmed death-ligand 1 expression in hepatocellular carcinoma. Histopathology. 2021;79:36–46.CrossRefPubMed Montasser A, et al. Transarterial chemoembolisation enhances programmed death-1 and programmed death-ligand 1 expression in hepatocellular carcinoma. Histopathology. 2021;79:36–46.CrossRefPubMed
18.
Zurück zum Zitat Sun B, et al. Safety and efficacy of lenvatinib combined with camrelizumab plus transcatheter arterial chemoembolization for unresectable hepatocellular carcinoma: a two-center retrospective study. Front Oncol. 2022;12: 982948.CrossRefPubMedPubMedCentral Sun B, et al. Safety and efficacy of lenvatinib combined with camrelizumab plus transcatheter arterial chemoembolization for unresectable hepatocellular carcinoma: a two-center retrospective study. Front Oncol. 2022;12: 982948.CrossRefPubMedPubMedCentral
19.
Zurück zum Zitat Cai M, et al. Transarterial chemoembolization combined with lenvatinib Plus PD-1 inhibitor for advanced hepatocellular carcinoma: a retrospective cohort study. Front Immunol. 2022;13: 848387.CrossRefPubMedPubMedCentral Cai M, et al. Transarterial chemoembolization combined with lenvatinib Plus PD-1 inhibitor for advanced hepatocellular carcinoma: a retrospective cohort study. Front Immunol. 2022;13: 848387.CrossRefPubMedPubMedCentral
20.
Zurück zum Zitat He M-K, et al. Lenvatinib, toripalimab, plus hepatic arterial infusion chemotherapy versus lenvatinib alone for advanced hepatocellular carcinoma. Ther Adv Med Oncol. 2021;13:17588359211002720.CrossRefPubMedPubMedCentral He M-K, et al. Lenvatinib, toripalimab, plus hepatic arterial infusion chemotherapy versus lenvatinib alone for advanced hepatocellular carcinoma. Ther Adv Med Oncol. 2021;13:17588359211002720.CrossRefPubMedPubMedCentral
21.
Zurück zum Zitat Hack SP, et al. IMbrave 050: a Phase III trial of atezolizumab plus bevacizumab in high-risk hepatocellular carcinoma after curative resection or ablation. Future Oncol. 2020;16:975–89.CrossRefPubMed Hack SP, et al. IMbrave 050: a Phase III trial of atezolizumab plus bevacizumab in high-risk hepatocellular carcinoma after curative resection or ablation. Future Oncol. 2020;16:975–89.CrossRefPubMed
22.
Zurück zum Zitat Ren Z, et al. Sintilimab plus a bevacizumab biosimilar (IBI305) versus sorafenib in unresectable hepatocellular carcinoma (ORIENT-32): a randomised, open-label, phase 2–3 study. Lancet Oncol. 2021;22:977–90.CrossRefPubMed Ren Z, et al. Sintilimab plus a bevacizumab biosimilar (IBI305) versus sorafenib in unresectable hepatocellular carcinoma (ORIENT-32): a randomised, open-label, phase 2–3 study. Lancet Oncol. 2021;22:977–90.CrossRefPubMed
24.
Zurück zum Zitat Zhu H-D, et al. Transarterial chemoembolization with PD-(L)1 inhibitors plus molecular targeted therapies for hepatocellular carcinoma (CHANCE001). Signal Transduct Target Ther. 2023;8:58.CrossRefPubMedPubMedCentral Zhu H-D, et al. Transarterial chemoembolization with PD-(L)1 inhibitors plus molecular targeted therapies for hepatocellular carcinoma (CHANCE001). Signal Transduct Target Ther. 2023;8:58.CrossRefPubMedPubMedCentral
27.
Zurück zum Zitat Cheng A-L, et al. Updated efficacy and safety data from IMbrave150: Atezolizumab plus bevacizumab vs. sorafenib for unresectable hepatocellular carcinoma. J Hepatol. 2022;76:862–73.CrossRefPubMed Cheng A-L, et al. Updated efficacy and safety data from IMbrave150: Atezolizumab plus bevacizumab vs. sorafenib for unresectable hepatocellular carcinoma. J Hepatol. 2022;76:862–73.CrossRefPubMed
28.
Zurück zum Zitat Anstee QM, Reeves HL, Kotsiliti E, Govaere O, Heikenwalder M. From NASH to HCC: current concepts and future challenges. Nat Rev Gastroenterol Hepatol. 2019;16:411–28.CrossRefPubMed Anstee QM, Reeves HL, Kotsiliti E, Govaere O, Heikenwalder M. From NASH to HCC: current concepts and future challenges. Nat Rev Gastroenterol Hepatol. 2019;16:411–28.CrossRefPubMed
29.
Zurück zum Zitat Pinato DJ, et al. Trans-arterial chemoembolization as a loco-regional inducer of immunogenic cell death in hepatocellular carcinoma: implications for immunotherapy. J Immunother Cancer. 2021;9: e003311.CrossRefPubMedPubMedCentral Pinato DJ, et al. Trans-arterial chemoembolization as a loco-regional inducer of immunogenic cell death in hepatocellular carcinoma: implications for immunotherapy. J Immunother Cancer. 2021;9: e003311.CrossRefPubMedPubMedCentral
31.
Zurück zum Zitat Huang Y, et al. Vascular normalizing doses of antiangiogenic treatment reprogram the immunosuppressive tumor microenvironment and enhance immunotherapy. Proc Natl Acad Sci USA. 2012;109:17561–6.ADSCrossRefPubMedPubMedCentral Huang Y, et al. Vascular normalizing doses of antiangiogenic treatment reprogram the immunosuppressive tumor microenvironment and enhance immunotherapy. Proc Natl Acad Sci USA. 2012;109:17561–6.ADSCrossRefPubMedPubMedCentral
32.
Zurück zum Zitat Palmer DH, Malagari K, Kulik LM. Role of locoregional therapies in the wake of systemic therapy. J Hepatol. 2020;72:277–87.CrossRefPubMed Palmer DH, Malagari K, Kulik LM. Role of locoregional therapies in the wake of systemic therapy. J Hepatol. 2020;72:277–87.CrossRefPubMed
33.
Zurück zum Zitat Wang W, Zhao Y, Bai W, et al. Response assessment for HCC patients treated with repeated TACE: the optimal time-point is still an open issue. J Hepatol. 2015;63(6):1530–1.CrossRefPubMed Wang W, Zhao Y, Bai W, et al. Response assessment for HCC patients treated with repeated TACE: the optimal time-point is still an open issue. J Hepatol. 2015;63(6):1530–1.CrossRefPubMed
34.
Zurück zum Zitat Hiroishi K, et al. Strong CD8+ T-cell responses against tumor-associated antigens prolong the recurrence-free interval after tumor treatment in patients with hepatocellular carcinoma. J Gastroenterol. 2010;45:451–8.CrossRefPubMed Hiroishi K, et al. Strong CD8+ T-cell responses against tumor-associated antigens prolong the recurrence-free interval after tumor treatment in patients with hepatocellular carcinoma. J Gastroenterol. 2010;45:451–8.CrossRefPubMed
35.
Zurück zum Zitat Zeng F, et al. Employing hypoxia characterization to predict tumour immune microenvironment, treatment sensitivity and prognosis in hepatocellular carcinoma. Comput Struct Biotechnol J. 2021;19:2775–89.CrossRefPubMedPubMedCentral Zeng F, et al. Employing hypoxia characterization to predict tumour immune microenvironment, treatment sensitivity and prognosis in hepatocellular carcinoma. Comput Struct Biotechnol J. 2021;19:2775–89.CrossRefPubMedPubMedCentral
36.
Zurück zum Zitat Kudo M, et al. Randomised, multicentre prospective trial of transarterial chemoembolisation (TACE) plus sorafenib as compared with TACE alone in patients with hepatocellular carcinoma: TACTICS trial. Gut. 2020;69:1492–501.CrossRefPubMed Kudo M, et al. Randomised, multicentre prospective trial of transarterial chemoembolisation (TACE) plus sorafenib as compared with TACE alone in patients with hepatocellular carcinoma: TACTICS trial. Gut. 2020;69:1492–501.CrossRefPubMed
37.
Zurück zum Zitat Ding X, et al. Transarterial chemoembolization plus lenvatinib versus transarterial chemoembolization plus sorafenib as first-line treatment for hepatocellular carcinoma with portal vein tumor thrombus: a prospective randomized study. Cancer. 2021;127:3782–93.CrossRefPubMed Ding X, et al. Transarterial chemoembolization plus lenvatinib versus transarterial chemoembolization plus sorafenib as first-line treatment for hepatocellular carcinoma with portal vein tumor thrombus: a prospective randomized study. Cancer. 2021;127:3782–93.CrossRefPubMed
Metadaten
Titel
Transarterial chemoembolization with/without immune checkpoint inhibitors plus tyrosine kinase inhibitors for unresectable hepatocellular carcinoma: a single center, propensity score matching real-world study
verfasst von
Guosheng Yuan
Wenli Li
Mengya Zang
Rong Li
Qi Li
Xiaoyun Hu
Qi Zhang
Wei Huang
Jian Ruan
Huajin Pang
Jinzhang Chen
Publikationsdatum
01.12.2024
Verlag
Springer US
Erschienen in
Discover Oncology / Ausgabe 1/2024
Print ISSN: 1868-8497
Elektronische ISSN: 2730-6011
DOI
https://doi.org/10.1007/s12672-024-00917-1

Weitere Artikel der Ausgabe 1/2024

Discover Oncology 1/2024 Zur Ausgabe

Alphablocker schützt vor Miktionsproblemen nach der Biopsie

16.05.2024 alpha-1-Rezeptorantagonisten Nachrichten

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

Mammakarzinom: Senken Statine das krebsbedingte Sterberisiko?

15.05.2024 Mammakarzinom Nachrichten

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

Labor, CT-Anthropometrie zeigen Risiko für Pankreaskrebs

13.05.2024 Pankreaskarzinom Nachrichten

Gerade bei aggressiven Malignomen wie dem duktalen Adenokarzinom des Pankreas könnte Früherkennung die Therapiechancen verbessern. Noch jedoch klafft hier eine Lücke. Ein Studienteam hat einen Weg gesucht, sie zu schließen.

Viel pflanzliche Nahrung, seltener Prostata-Ca.-Progression

12.05.2024 Prostatakarzinom Nachrichten

Ein hoher Anteil pflanzlicher Nahrung trägt möglicherweise dazu bei, das Progressionsrisiko von Männern mit Prostatakarzinomen zu senken. In einer US-Studie war das Risiko bei ausgeprägter pflanzlicher Ernährung in etwa halbiert.

Update Onkologie

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